US9359360B2 - TLR agonists - Google Patents

TLR agonists Download PDF

Info

Publication number
US9359360B2
US9359360B2 US13/682,208 US201213682208A US9359360B2 US 9359360 B2 US9359360 B2 US 9359360B2 US 201213682208 A US201213682208 A US 201213682208A US 9359360 B2 US9359360 B2 US 9359360B2
Authority
US
United States
Prior art keywords
compound
alkyl
formula
compounds
tlr
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US13/682,208
Other versions
US20130165455A1 (en
Inventor
Dennis A. Carson
Kenji Takabayashi
Howard B. Cottam
Michael Chan
Christina C. N. Wu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of California
Original Assignee
University of California
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of California filed Critical University of California
Priority to US13/682,208 priority Critical patent/US9359360B2/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: UNIVERSITY OF CALIFORNIA SAN DIEGO
Publication of US20130165455A1 publication Critical patent/US20130165455A1/en
Assigned to THE REGENTS OF THE UNIVERSITY OF CALIFORNIA reassignment THE REGENTS OF THE UNIVERSITY OF CALIFORNIA ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GRIMSHAW, SUZANNE, CARSON, DENNIS A, CHAN, MICHAEL, COTTAM, HOWARD B., WU, CHRISTINA C.
Application granted granted Critical
Publication of US9359360B2 publication Critical patent/US9359360B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/16Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 two nitrogen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/18Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 one oxygen and one nitrogen atom, e.g. guanine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/02Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6
    • C07D473/24Heterocyclic compounds containing purine ring systems with oxygen, sulphur, or nitrogen atoms directly attached in positions 2 and 6 one nitrogen and one sulfur atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/26Heterocyclic compounds containing purine ring systems with an oxygen, sulphur, or nitrogen atom directly attached in position 2 or 6, but not in both
    • C07D473/32Nitrogen atom
    • C07D473/34Nitrogen atom attached in position 6, e.g. adenine

Definitions

  • PAMPs pathogen associated molecular patterns
  • PAMPs include peptidoglycans, lipotechoic acids from gram-positive cell walls, the sugar mannose (which is common in microbial carbohydrates but rare in humans), bacterial DNA, double-stranded RNA from viruses, and glucans from fungal cell walls.
  • TLRs Toll-like Receptors
  • TLR7 and TLR9 recognize and respond to imiquimod and immunostimulatory CpG oligonucleotides (ISS-ODN), respectively.
  • the synthetic immunomodulator R-848 activates both TLR7 and TLR8. While TLR stimulation initiates a common signaling cascade (involving the adaptor protein MyD88, the transcription factor NF-kB, and pro-inflammatory and effector cytokines), certain cell types tend to produce certain TLRs. For example, TLR7 and TLR9 are found predominantly on the internal faces of endosomes in dendritic cells (DCs) and B lymphocytes (in humans; mouse macrophages express TLR7 and TLR9). TLR8, on the other hand, is found in human blood monocytes. (See Hornung et al., J Immunol, 168:4531-4537 (2002)).
  • Interferons are also involved in the efficient induction of an immune response, especially after viral infection (Brassard et al., J. Leukoc Biol, 71:568-581 (2002).) However, many viruses produce a variety of proteins that block interferon production or action at various levels. Antagonism of interferon is believed to be part of a general strategy to evade innate, as well as adaptive immunity. (See Levy et al., Cytokine Growth Factor Rev, 12:143-156 (2001).) While TLR agonists (TLR-L) may be sufficiently active for some methods of treatment, in some instances the microbial interferon antagonists could mitigate the adjuvant effects of synthetic TLR-L.
  • the present invention provides for TLR agonist conjugates (compounds) and compositions, as well as methods of using them.
  • the compounds of the invention are broad-spectrum, long-lasting, and non-toxic combination of synthetic immunostimulatory agents, which are useful for activating the immune system of a mammal, preferably a human and can help direct the pharmacophore to the receptor within the endosomes of target cells and enhance the signal transduction induced by the pharmacophore.
  • the compounds of the invention include a pharmacophore covalently bound to an auxiliary group. Accordingly there is provided a compound of the invention which is a compound of formula (I):
  • X 1 is —O—, —S—, or —NR c —;
  • R c hydrogen, C 1-10 alkyl, or C 1-10 alkyl substituted by C 3-6 -cycloalkyl, or R c and R 1 taken together with the nitrogen atom can form a heterocyclic ring or a substituted heterocyclic ring, wherein the substituents are hydroxy, C 1-6 alkyl, hydroxyC 1-6 alkylene, C 1-6 alkoxy, C 1-6 alkoxyC 1-6 alkylene, or cyano;
  • R 1 is (C 1 -C 10 )alkyl, substituted (C 1 -C 10 )alkyl, C 6-10 aryl, or substituted C 6-10 aryl, C 5-9 heterocyclic, substituted C 5-9 heterocyclic;
  • each R 2 is independently hydrogen, —OH, (C 1 -C 6 )alkyl, substituted (C 1 -C 6 )alkyl, (C 1 -C 6 )alkoxy, substituted (C 1 -C 6 )alkoxy, —C(O)—(C 1 -C 6 )alkyl (alkanoyl), substituted —C(O)—(C 1 -C 6 )alkyl, —C(O)—(C 6 -C 10 )aryl (aroyl), substituted —C(O)—(C 6 -C 10 )aryl, —C(O)OH (carboxyl), —C(O)O(C 1 -C 6 )alkyl (alkoxycarbonyl), substituted —C(O)O(C 1 -C 6 )alkyl, —NR a R b , —C(O)NR a R b (carbamoyl), substituted
  • each R a and R b is independently hydrogen, (C 1 -C 6 )alkyl, (C 3 -C 8 )cycloalkyl, (C 1 -C 6 )alkoxy, halo(C 1 -C 6 )alkyl, (C 3 -C 8 )cycloalkyl(C 1 -C 6 )alkyl, (C 1 -C 6 )alkanoyl, hydroxy(C 1 -C 6 )alkyl, aryl, aryl(C 1 -C 6 )alkyl, Het, Het (C 1 -C 6 )alkyl, or (C 1 -C 6 )alkoxycarbonyl;
  • X 2 is a bond or a linking group; and R 3 is an auxiliary group;
  • the auxiliary groups can include organic molecules, composed of carbon, oxygen, hydrogen, nitrogen, sulfur, phosphorus atoms. These groups are not harmful to body tissues (e.g., they are non-toxic, and/or do not cause inflammation).
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising at least one compound of formula (I), or a pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable diluent or carrier.
  • the invention provides a therapeutic method for preventing or treating a pathological condition or symptom in a mammal, such as a human, wherein the activity of TLR agonists is implicated and its action is desired, comprising administering to a mammal in need of such therapy, an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof.
  • pathological conditions or symptoms that are suitable for treatment include cancers, treatment for bacterial or viral diseases, treating autoimmune diseases, and treating Crohn's Disease.
  • the compounds of the invention can also be used as or to prepare vaccines against bacteria, viruses, cancer cells, cancer specific peptides, enhancers of monoclonal antibodies against cancer, a CNS stimulant, or for biodefense.
  • the invention provides a compound of formula (I) for use in medical therapy (e.g., for use as an anti-cancer agent, treatment for bacterial diseases, treatment for viral diseases, such as hepatitis C and hepatitis B, Crohn's Disease, and as therapeutic agents for treating immunologic disease). Furthermore, it is suggested that compounds of formula (I) will prevent carcinogenesis by hepatitis C and hepatitis B, as well as the use of a compound of formula (I) for the manufacture of a medicament useful for the treatment of cancer, viral diseases, Crohn's Disease, and immunologic disorders in a mammal, such as a human.
  • medical therapy e.g., for use as an anti-cancer agent, treatment for bacterial diseases, treatment for viral diseases, such as hepatitis C and hepatitis B, Crohn's Disease, and as therapeutic agents for treating immunologic disease.
  • compounds of formula (I) will prevent carcinogenesis by hepatitis C and hepatitis B, as well
  • the present invention provides a method for treating a viral infection in a mammal by administering a TLR agonist compound of formula (I).
  • the viral infection can be caused by an RNA virus, a product of the RNA virus that acts as a TLR agonist and/or a DNA virus.
  • a specific DNA virus for treatment is the Hepatitis B virus.
  • the present invention provides a method for treating cancer by administering an effective amount of a TLR agonist compound of formula (I).
  • the cancer can be an interferon sensitive cancer, such as, for example, a leukemia, a lymphoma, a myeloma, a melanoma, or a renal cancer.
  • the present invention provides a method of treating an autoimmune disease by administering a therapeutically effective amount of a TLR agonist compound of formula (I) or a pharmaceutically acceptable salt of such a compound.
  • a specific autoimmune disease is Multiple Sclerosis, lupus, rheumatoid arthritis and the like.
  • the present invention provides a method of treating Crohn's Disease by administering a TLR agonist compound of formula (I).
  • the TLR agonists can be a homofunctional TLR agonist polymer and can consist of a TLR-7 agonist or a TLR-8 agonist.
  • the TLR7 agonist can be a 7-thia-8-oxoguanosinyl (TOG) moiety, a 7-deazaguanosinyl (7DG) moiety, a resiquimod moiety, or an imiquimod moiety.
  • the TLR8 agonist can be a resiquimod moiety.
  • the TLR agonist is a heterofunctional TLR agonist polymer.
  • the heterofunctional TLR agonist polymer can include a TLR-7 agonist and a TLR-8 agonist or a TLR-9 agonist or all three agonists.
  • the heterofunctional TLR agonist polymer can include a TLR-8 agonist and a TLR-9 agonist.
  • the invention also provides processes and intermediates disclosed herein that are useful for preparing compounds of formula (I) or salts thereof.
  • FIG. 1 is a graphic illustration of the absorption chromophore (at ⁇ 350 nm) of a compound of formula I (OVA/IV150 Conjugate).
  • FIG. 2 is a graphic illustration of the stimulation of bone marrow derived dendritic cells (BMDC).
  • FIG. 3 illustrates the conjugation of a TLR7 agonist, UC-1V150, to mouse serum albumin (MSA). The success of conjugation is indicated by UV spectroscopy.
  • the UC-1V150 to MSA ratio is approximately 5:1
  • FIGS. 4A and B illustrate that the UC-1V150 and MSA conjugates activate both murine bone marrow-derived macrophages ( 4 A) and human peripheral blood mononuclear cells ( 4 B).
  • Cells were incubated with various concentrations of the compound from 0.5 nM to 10 ⁇ M in BMDM or from 0.1 to 10 ⁇ M in PBMC. Culture supernatants were harvested after 24 h and cytokine levels were analyzed by Luminex.
  • FIGS. 5A, 5B, 5C, and 5D illustrate the increased potency and duration of effect of UC-1V150/MSA.
  • C57BL/6 mice were injected (i.v.) with (A) 0.1 micromole of SM-360320, a TLR7 ligand, or (B) equivalent amount of a TLR7 agonist UC-1V150 (aldehyde-modified SM-360320) or UC-1V150/MSA to 500 ⁇ g MSA per mouse.
  • Serum samples were collected at the indicated time points and cytokine levels were analyzed by Luminex.
  • MSA mouse serum albumin.
  • the effect from the original TLR7 ligand, SM-360320 lasted for only 2 hours whereas UC-1V150/MSA has extended the effect to at least 6 hours.
  • FIG. 6 illustrates the effects of UC-1V150 conjugated with inactivated SIV ( 6 A) or with OVA in combination with ODN ( 6 B).
  • Myeloid BMDC were incubated for 24 hr with various conditions at 0.1 ⁇ g/ml as indicated.
  • IL-12 levels in the cell supernatant were measured by ELISA.
  • FIGS. 7A and 7B illustrates an increased potency of UC-1V150/MSA.
  • C57BL/6 mice were i.v. injected with 380 nmole of SM-360320 or UC-1V150, or 500 ⁇ g of UC-1V150/MSA (equivalent to 3.8 nmole UC-1V150) per mouse.
  • Serum samples were collected after 2 h and cytokine levels were analyzed by Luminex. To achieve the similar effect, at least 100-fold higher concentration of either UC-1V150 or SM-360320 was required as compared to that of UC-1V150/MSA.
  • FIG. 8 is an illustration of the uv spectrum of a double-conjugate, (OVA/IV150/1043).
  • FIG. 9 is an illustration of the induction of IL-12 in BMDC using OVA/ODN/IV150 conjugates.
  • FIG. 10 illustrates direct conjugation of SIV Particles to the IA compound IV150.
  • FIG. 11 illustrates the ability to prepare compounds of the invention with virus particles attached to a compound having formula IA and the TLR agonist activity of the compounds.
  • FIG. 12 illustrates the molecular areas of specificity for antibodies raised against the conjugates containing a linker and a TLR ligand.
  • FIGS. 13A and 13B illustrate the distinction between the four substances applied to the respective lanes on a gel in a Western plot analysis.
  • the gel membrane was probed with anti-ovalbumin (anti-OVA) antibody and all lanes gave a positive band, indicating that OVA was detected in all lanes, as expected.
  • the gel membrane was probed with the selective antibody raised to the TLR ligand portion of the conjugate, and therefore only lane 4 was positive, confirming the specificity of the antibody for the TLR ligand.
  • auxiliary groups include side chains that increase solubility, such as, for example, groups containing morpholino, piperidino, pyrrolidino, or piperazino rings and the like; amino acids, polymers of amino acids (proteins or peptides), e.g., dipeptides or tripeptides, and the like; carbohydrates (polysaccharides), nucleotides such as, for example, PNA, RNA and DNA, and the like; polymers of organic materials, such as, for example, polyethylene glycol, poly-lactide and the like; monomeric and polymeric lipids; insoluble organic nanoparticles; non-toxic body substances such as, for example, cells, lipids, vitamins, co-factors, antigens such as, for example microbes, such as, for example, viruses, bacteria, fungi, and the like.
  • the antigens can include inactivated whole organisms, or sub-components thereof and the like.
  • the compounds of the invention can be prepared using compounds having formula (IA):
  • X is a group that can react to form a bond to the linking group or can react to form a bond to the auxiliary group.
  • a specific group of compounds having formula (IA) are disclosed in U.S. Pat. No. 6,329,381.
  • halo is fluoro, chloro, bromo, or iodo.
  • Alkyl, alkoxy, alkenyl, alkynyl, etc. denote both straight and branched groups; but reference to an individual radical such as “propyl” embraces only the straight chain radical, a branched chain isomer such as “isopropyl” being specifically referred to.
  • Aryl denotes a phenyl radical or an ortho-fused bicyclic carbocyclic radical having about nine to ten ring atoms in which at least one ring is aromatic.
  • Heteroaryl encompasses a radical attached via a ring carbon of a monocyclic aromatic ring containing five or six ring atoms consisting of carbon and one to four heteroatoms each selected from the group consisting of non-peroxide oxygen, sulfur, and N(X) wherein X is absent or is H, O, (C 1 -C 4 )alkyl, phenyl or benzyl, as well as a radical of an ortho-fused bicyclic heterocycle of about eight to ten ring atoms derived therefrom, particularly a benz-derivative or one derived by fusing a propylene, trimethylene, or tetramethylene diradical thereto.
  • Processes for preparing compounds of formula I or for preparing intermediates useful for preparing compounds of formula I are provided as further embodiments of the invention.
  • Intermediates useful for preparing compounds of formula I are also provided as further embodiments of the invention.
  • salts may be appropriate.
  • acceptable salts are organic acid addition salts formed with acids which form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, ⁇ -ketoglutarate, and ⁇ -glycerophosphate.
  • Suitable inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts.
  • Acceptable salts may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion.
  • a sufficiently basic compound such as an amine
  • a suitable acid affording a physiologically acceptable anion.
  • Alkali metal (for example, sodium, potassium or lithium) or alkaline earth metal (for example calcium) salts of carboxylic acids can also be made.
  • Alkyl includes straight or branched C 1-10 alkyl groups, e.g., methyl, ethyl, propyl, butyl, pentyl, isopropyl, isobutyl, 1-methylpropyl, 3-methylbutyl, hexyl, and the like.
  • Lower alkyl includes straight or branched C 1-6 alkyl groups, e.g., methyl, ethyl, propyl, 1-methylethyl, butyl, 1-methylpropyl, 2-methylpropyl, 1,1-dimethylethyl, pentyl, 1-methylbutyl, 2-methylbutyl, 3-methylbutyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, and the like.
  • C 1-6 alkyl groups e.g., methyl, ethyl, propyl, 1-methylethyl, butyl, 1-methylpropyl, 2-methylpropyl, 1,1-dimethylethyl, pentyl, 1-methylbutyl, 2-methylbutyl, 3-methylbutyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, and the like.
  • alkylene refers to a divalent straight or branched hydrocarbon chain (e.g. ethylene —CH 2 —CH 2 —).
  • Cycloalkyl includes groups such as, cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, and the like, and alkyl-substituted C 3-7 cycloalkyl group, preferably straight or branched C 1-6 alkyl group such as methyl, ethyl, propyl, butyl or pentyl, and C 5-7 cycloalkyl group such as, cyclopentyl or cyclohexyl, and the like.
  • Lower alkoxy includes C 1-6 alkoxy groups, such as methoxy, ethoxy or propoxy, and the like.
  • Lower alkanoyl includes C 1-6 alkanoyl groups, such as formyl, acetyl, propanoyl, butanoyl, pentanoyl or hexanoyl, and the like.
  • C 7-11 aroyl includes groups such as benzoyl or naphthoyl;
  • Lower alkoxycarbonyl includes C 2-7 alkoxycarbonyl groups, such as methoxycarbonyl, ethoxycarbonyl or propoxycarbonyl, and the like.
  • Lower alkylamino group means amino group substituted by C 1-6 alkyl group, such as, methylamino, ethylamino, propylamino, butylamino, and the like.
  • Di(lower alkyl)amino group means amino group substituted by the same or different and C 1-6 alkyl group (e.g. dimethylamino, diethylamino, ethylmethylamino).
  • Lower alkylcarbamoyl group means carbamoyl group substituted by C 1-6 alkyl group (e.g. methylcarbamoyl, ethylcarbamoyl, propylcarbamoyl, butylcarbamoyl).
  • Di(lower alkyl)carbamoyl group means carbamoyl group substituted by the same or different and C 1-6 alkyl group (e.g. dimethylcarbamoyl, diethylcarbamoyl, ethylmethylcarbamoyl).
  • Halogen atom means halogen atom such as fluorine atom, chlorine atom, bromine atom or iodine atom.
  • Aryl refers to a C 6-10 monocyclic or fused cyclic aryl group, such as phenyl, indenyl, or naphthyl, and the like.
  • Heterocyclic refers to monocyclic saturated heterocyclic groups, or unsaturated monocyclic or fused heterocyclic group containing at least one heteroatom, e.g., 0-3 nitrogen atoms (—NR d —), 0-1 oxygen atom (—O—), and 0-1 sulfur atom (—S—).
  • saturated monocyclic heterocyclic group includes 5 or 6 membered saturated heterocyclic group, such as tetrahydrofuranyl, pyrrolidinyl, morpholinyl, piperidyl, piperazinyl or pyrazolidinyl.
  • Non-limiting examples of unsaturated monocyclic heterocyclic group includes 5 or 6 membered unsaturated heterocyclic group, such as furyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, pyridyl or pyrimidinyl.
  • Non-limiting examples of unsaturated fused heterocyclic groups includes unsaturated bicyclic heterocyclic group, such as indolyl, isoindolyl, quinolyl, benzothizolyl, chromanyl, benzofuranyl, and the like.
  • heterocyclic rings include 5 or 6 membered saturated heterocyclic rings, such as 1-pyrrolidinyl, 4-morpholinyl, 1-piperidyl, 1-piperazinyl or 1-pyrazolidinyl, 5 or 6 membered unsaturated heterocyclic rings such as 1-imidazolyl, and the like.
  • the alkyl, aryl, heterocyclic groups of R 1 can be optionally substituted with one or more substituents, wherein the substituents are the same or different, and include lower alkyl; cycloalkyl, hydroxyl; hydroxy C 1-6 alkylene, such as hydroxymethyl, 2-hydroxyethyl or 3-hydroxypropyl; lower alkoxy; C 1-6 alkoxy C 1-6 alkyl, such as 2-methoxyethyl, 2-ethoxyethyl or 3-methoxypropyl; amino; alkylamino; dialkyl amino; cyano; nitro; acyl; carboxyl; lower alkoxycarbonyl; halogen; mercapto; C 1-6 alkylthio, such as, methylthio, ethylthio, propylthio or butylthio; substituted C 1-6 alkylthio, such as methoxyethylthio, methylthioethylthio, hydroxyeth
  • the alkyl, aryl, heterocyclic groups of R 2 can be optionally substituted with one or more substituents, wherein the substituents are the same or different, and include hydroxyl; C 1-6 alkoxy, such as methoxy, ethoxy or propoxy; carboxyl; C 2-7 alkoxycarbonyl, such as methoxycarbonyl, ethoxycarbonyl or propoxycarbonyl) and halogen.
  • the alkyl, aryl, heterocyclic groups of R c can be optionally substituted with one or more substituents, wherein the substituents are the same or different, and include C 3-6 cycloalkyl; hydroxyl; C 1-6 alkoxy; amino; cyano; aryl; substituted aryl, such as 4-hydroxyphenyl, 4-methoxyphenyl, 4-chlorophenyl or 3,4-dichlorophenyl; nitro and halogen.
  • the heterocyclic ring formed together with R c and R 1 and the nitrogen atom to which they are attached can be optionally substituted with one or more substituents, wherein the substituents are the same or different, and include C 1-6 alkyl; hydroxy C 1-6 alkylene; C 1-6 alkoxy C 1-6 alkylene; hydroxyl; C 1-6 alkoxy; and cyano.
  • amino acid as used herein, comprises the residues of the natural amino acids (e.g. Ala, Arg, Asn, Asp, Cys, Glu, Gln, Gly, His, Hyl, Hyp, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, and Val) in D or L form, as well as unnatural amino acids (e.g.
  • the term also comprises natural and unnatural amino acids bearing a conventional amino protecting group (e.g.
  • acetyl or benzyloxycarbonyl as well as natural and unnatural amino acids protected at the carboxy terminus (e.g. as a (C 1 -C 6 )alkyl, phenyl or benzyl ester or amide; or as an -methylbenzyl amide).
  • suitable amino and carboxy protecting groups are known to those skilled in the art (See for example, T. W. Greene, Protecting Groups In Organic Synthesis ; Wiley: New York, 1981, and references cited therein).
  • An amino acid can be linked to the remainder of a compound of formula I through the carboxy terminus, the amino terminus, or through any other convenient point of attachment, such as, for example, through the sulfur of cysteine.
  • TLR toll-like receptor
  • PAMPs pathogen associated molecular patterns
  • TLR agonist refers to a molecule that binds to a TLR and antagonizes the receptor.
  • Synthetic TLR agonists are chemical compounds that are designed to bind to a TLR and activate the receptor.
  • Exemplary novel TLR agonists provided herein include “TLR-7 agonist” “TLR-8 agonist” and “TLR-9 agonist.”
  • nucleic acid refers to DNA, RNA, single-stranded, double-stranded, or more highly aggregated hybridization motifs, and any chemical modifications thereof. Modifications include, but are not limited to, those providing chemical groups that incorporate additional charge, polarizability, hydrogen bonding, electrostatic interaction, and fluxionality to the nucleic acid ligand bases or to the nucleic acid ligand as a whole.
  • Such modifications include, but are not limited to, peptide nucleic acids (PNAs), phosphodiester group modifications (e.g., phosphorothioates, methylphosphonates), 2′-position sugar modifications, 5-position pyrimidine modifications, 7-position purine modifications, 8-position purine modifications, 9-position purine modifications, modifications at exocyclic amines, substitution of 4-thiouridine, substitution of 5-bromo or 5-iodo-uracil; backbone modifications, methylations, unusual base-pairing combinations such as the isobases, isocytidine and isoguanidine and the like.
  • Nucleic acids can also include non-natural bases, such as, for example, nitroindole. Modifications can also include 3′ and 5′ modifications such as capping with a BHQ, a fluorophore or another moiety.
  • a specific value for X 1 is a sulfur atom, an oxygen atom or —NR c —.
  • Another specific X 1 is a sulfur atom.
  • Another specific X 1 is an oxygen atom.
  • Another specific X 1 is —NR c —.
  • Another specific X 1 is —NH—.
  • R c is hydrogen, C 1-4 alkyl or substituted C 1-4 alkyl.
  • R 1 and R c taken together is when they form a heterocyclic ring or a substituted heterocyclic ring.
  • R 1 and R c taken together is substituted or unsubstituted morpholino, piperidino, pyrrolidino, or piperazino ring.
  • R 1 is hydrogen, C 1-4 alkyl, or substituted C 1-4 alkyl.
  • R 1 is 2-hydroxyethyl, 3-hydroxypropyl, 4-hydroxybutyl, 2-aminoethyl, 3-aminopropyl, 4-aminobutyl, methoxymethyl, 2-methoxyethyl, 3-methoxypropyl, ethoxymethyl, 2-ethoxyethyl, methylthiomethyl, 2-methylthioethyl, 3-methylthiopropyl, 2-fluoroethyl, 3-fluoropropyl, 2,2,2-trifluoroethyl, cyanomethyl, 2-cyanoethyl, 3-cyanopropyl, methoxycarbonylmethyl, 2-methoxycarbonylethyl, 3-methoxycarbonylpropyl, benzyl, phenethyl, 4-pyridylmethyl, cyclohexylmethyl, 2-thienylmethyl, 4-methoxyphenylmethyl, 4-hydroxyphenylmethyl, 4-fluorophenylmethyl, or 4-chlorophen
  • R 1 is hydrogen, CH 3 —, CH 3 —CH 2 —, CH 3 CH 2 CH 2 —, hydroxyC 1-4 alkylene, or C 1-4 alkoxyC 1-4 alkylene.
  • R 1 Another specific value for R 1 is hydrogen, CH 3 —, CH 3 —CH 2 —, CH 3 —O—CH 2 CH 2 — or CH 3 —CH 2 —O—CH 2 CH 2 —.
  • R 2 is hydrogen, halogen, or C 1-4 alkyl.
  • R 2 is hydrogen, chloro, bromo, CH 3 —, or CH 3 —CH 2 —.
  • substituents for substitution on the alkyl, aryl or heterocyclic groups are hydroxy, C 1-6 alkyl, hydroxyC 1-6 alkylene, C 1-6 alkoxy, C 1-6 alkoxyC 1-6 alkylene, C 3-6 cycloalkyl, amino, cyano, halogen, or aryl.
  • a specific value for X 2 is a bond or a chain having up to about 24 atoms; wherein the atoms are selected from the group consisting of carbon, nitrogen, sulfur, non-peroxide oxygen, and phosphorus.
  • X 2 is a bond or a chain having from about 4 to about 12 atoms.
  • X 2 is a bond or a chain having from about 6 to about 9 atoms.
  • a specific auxiliary group is an amino acid, a carbohydrate, a peptide, an antigen, a nucleic acid, a body substance, or a microbe.
  • a specific peptide has from 2 to about 20 amino acid residues.
  • Another specific peptide has from 10 to about 20 amino acid residues.
  • a specific auxiliary group is a carbohydrate.
  • a specific nucleic acid is DNA, RNA or PNA.
  • a specific body substance is a cell, lipid, vitamin, or co-factor.
  • Another specific body substance is a cell or lipid.
  • a specific antigen is a microbe.
  • a specific microbe is a virus, bacteria, or fungi.
  • Another specific microbe is a virus or a bacteria.
  • Bacillus anthracis anthrax
  • Listeria monocytogenes Listeria monocytogenes
  • Francisella tularensis Listeria monocytogenes
  • Salmonella Salmonella
  • Salmonella are typhimurium or enteritidis.
  • RNA viruses RNA viruses, a product of the RNA virus, or a DNA virus.
  • a specific DNA virus is the Hepatitis B virus.
  • Specific compounds of the invention have the general formula IA-L-A 1 ; IA-L-(A 1 ) 2 ; IA-L-A 1 -A 1 ; IA-L-A 1 -L-A 1 ; (IA) 2 -L-A 1 -A 1 ; (IA) 2 -L-A 1 -L-A 1 ; (IA) 2 -L-A 1 ; or (IA) 2 -L-(A 1 ) 2 ; wherein IA is as disclosed herein; L is absent or is a linking group; and each A 1 group independently represents an auxiliary group.
  • the viral infection is caused by a coronavirus that causes Severe Acute Respiratory Syndrome (SARS), a Hepatitis B virus, or a Hepatitis C Virus.
  • SARS Severe Acute Respiratory Syndrome
  • Hepatitis B virus a Hepatitis B virus
  • Hepatitis C Virus a Hepatitis C virus
  • the viral infection is caused by a coronavirus that causes Severe Acute Respiratory Syndrome (SARS), a Hepatitis B virus, or a Hepatitis C Virus.
  • SARS Severe Acute Respiratory Syndrome
  • Hepatitis B virus a Hepatitis B virus
  • Hepatitis C Virus a coronavirus that causes Severe Acute Respiratory Syndrome
  • Specific cancers that can be treated include melanoma, superficial bladder cancer, actinic keratoses, intraepithelial neoplasia, and basal cell skin carcinoma, squamous, and the like.
  • the method of the invention includes treatment for a precancerous condition such as, for example, actinic keratoses or intraepithelial neoplasia, familial polyposis (polyps), cervical dysplasia, cervical cancers, superficial bladder cancer, and any other cancers associated with infection (e.g., lymphoma Karposi's sarcoma, or leukemia); and the like.
  • Non limiting examples of the pathological conditions or symptoms that can be treated include viral diseases, cancer, inflammatory diseases of the gastrointestinal tract, brain, skin, joints, and other tissues.
  • the auxiliary groups are believed to enhance the drug activity of the pharmacophore (compounds of formula (I)) by (a) helping to direct the pharmacophore to the receptor within the endosomes of target cells; (b) by enhancing signal transduction induced by the pharmacophore, by cross-linking the receptor; and/or (c) the pharmacophore can enhance the response to the auxiliary group (e.g., immune response).
  • the auxiliary groups should form generally stable bonds with the pharmacophore, and do not act as prodrugs.
  • IMPDH inosine monophosphate dehydrogenase
  • an IMPDH inhibitor refers to an inhibitor of the enzyme inosine monophosphate dehydrogenase.
  • IMPDH inhibitors are used clinically: ribavirin, mizoribine, and mycophenolate mofetil.
  • Ribavirin and mizoribine are prodrugs that are phosphorylated intracellularly to produce IMP analogs (Goldstein et al., Cuff Med Chem, 6:519-536 (1999)). Viramidine is a prodrug of Ribavirin. Mycophenolate mofetil is immunosuppressive, and has gastrointestinal irritative properties that may be attributable to its enterohepatic recirculation (Papageorgiou C, Mini Rev Med Chem., 1:71-77 (2001)). Mizoribine aglycone, a prodrug, is used as an IMPDH inhibitor. Other non-limiting examples IMPDH inhibitors, including prodrugs of mizoribine and mizoribine aglycone are known and are disclosed in published U.S. Patent application No. 20050004144.
  • salts are organic acid addition salts formed with acids which form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, ⁇ -ketoglutarate, and ⁇ -glycerophosphate.
  • Suitable inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts.
  • salts may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion.
  • a sufficiently basic compound such as an amine
  • a suitable acid affording a physiologically acceptable anion.
  • Alkali metal (for example, sodium, potassium or lithium) or alkaline earth metal (for example calcium) salts of carboxylic acids can also be made.
  • the compounds (conjugates) of the invention can be prepared using standard synthetic methods known in the art.
  • a general ester synthesis is illustrated below:
  • the compounds of formula I can be formulated as pharmaceutical compositions and administered to a mammalian host, such as a human patient in a variety of forms adapted to the chosen route of administration, i.e., orally or parenterally, by intravenous, intramuscular, topical or subcutaneous routes.
  • the present compounds may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier. They may be enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet.
  • a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier.
  • the active compound may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • Such compositions and preparations should contain at least 0.1% of active compound.
  • the percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 60% of the weight of a given unit dosage form.
  • the amount of active compound in such therapeutically useful compositions is such that an effective dosage level will be obtained.
  • the tablets, troches, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added.
  • a liquid carrier such as a vegetable oil or a polyethylene glycol.
  • any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed.
  • the active compound may be incorporated into sustained-release preparations and devices.
  • the active compound may also be administered intravenously or intraperitoneally by infusion or injection.
  • Solutions of the active compound or its salts can be prepared in water, optionally mixed with a nontoxic surfactant.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes.
  • the ultimate dosage form should be sterile, fluid and stable under the conditions of manufacture and storage.
  • the liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compound in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization.
  • the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
  • the present compounds may be applied in pure form, i.e., when they are liquids. However, it will generally be desirable to administer them to the skin as compositions or formulations, in combination with a dermatologically acceptable carrier, which may be a solid or a liquid.
  • Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like.
  • Useful liquid carriers include water, alcohols or glycols or water-alcohol/glycol blends, in which the present compounds can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants.
  • Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use.
  • the resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers.
  • Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.
  • Examples of useful dermatological compositions which can be used to deliver the compounds of formula I to the skin are known to the art; for example, see Jacquet et al. (U.S. Pat. No. 4,608,392), Geria (U.S. Pat. No. 4,992,478), Smith et al. (U.S. Pat. No. 4,559,157) and Wortzman (U.S. Pat. No. 4,820,508).
  • Useful dosages of the compounds of formula I can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949.
  • the concentration of the compound(s) of formula I in a liquid composition will be from about 0.1-25 wt-%, preferably from about 0.5-10 wt-%.
  • concentration in a semi-solid or solid composition such as a gel or a powder will be about 0.1-5 wt-%, preferably about 0.5-2.5 wt-%.
  • the amount of the compound, or an active salt or derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician.
  • a suitable dose will be in the range of from about 0.5 to about 100 mg/kg, e.g., from about 10 to about 75 mg/kg of body weight per day, such as 3 to about 50 mg per kilogram body weight of the recipient per day, preferably in the range of 6 to 90 mg/kg/day, most preferably in the range of 15 to 60 mg/kg/day.
  • the compound is conveniently administered in unit dosage form; for example, containing 5 to 1000 mg, conveniently 10 to 750 mg, most conveniently, 50 to 500 mg of active ingredient per unit dosage form.
  • the active ingredient should be administered to achieve peak plasma concentrations of the active compound of from about 0.5 to about 75 ⁇ M, preferably, about 1 to 50 ⁇ M, most preferably, about 2 to about 30 ⁇ M. This may be achieved, for example, by the intravenous injection of a 0.05 to 5% solution of the active ingredient, optionally in saline, or orally administered as a bolus containing about 1-100 mg of the active ingredient. Desirable blood levels may be maintained by continuous infusion to provide about 0.01-5.0 mg/kg/hr or by intermittent infusions containing about 0.4-15 mg/kg of the active ingredient(s).
  • the desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day.
  • the sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.
  • TLR agonist The ability of a compound of the invention to act as a TLR agonist may be determined using pharmacological models which are well known to the art, including the procedures disclosed by Lee et al.; PNAS, 100 p 6646-6651, 2003.
  • Sodium salt of 2-methoxyethanol is generated by dissolving sodium metal (81 mg) in 2-methoxyethanol (30 mL) with heat. To this solution is added the product of example 2 (1.0 g) dissolved in methoxyethanol (300 mL, with heat). The reaction mixture is heated for 8 h at 115° C. bath temperature, concentrated in vacuo to near dryness and the residue partitioned between ethyl acetate and water. Flash silica gel chromatography of the organic layer using 5% methanol in dichloromethane gave 763 mg product. NMR is consistent with structure assignment.
  • This reducing agent used to convert the nitrile to the aldehyde function is prepared essentially as described in Bull. Korean Chem. Soc . (2002), 23(12), 1697-1698. A 0.5 M solution in dry THF is prepared.
  • example 5 The product of example 5 (100 mg) is dissolved in dry THF (3 mL) and cooled to 0° C. under argon.
  • the reagent generated in example 6 (0.72 mL) is added to the reaction flask and the mixture is stirred at 0-5° C. for 1 h and then quenched by addition of 3 M HCl.
  • the mixture is then extracted with ethyl acetate and then dichloromethane and concentrated in vacuo to yield 85 mg.
  • NMR is consistent with structure assignment.
  • Example 7 The product of example 7 (800 mg) is combined with sodium iodide (504 mg) and acetonitrile (40 mL), and then chlorotrimethylsilane (0.5 mL) is slowly added. The mixture is heated at 70° C. for 3.5 h, cooled and filtered. The solid product is washed with water, then ether to yield 406 mg. NMR, UV, MS are consistent with structure assignment. This material is suitable for conjugation reactions between linkers and auxiliary groups.

Abstract

The present invention provides for TLR agonist conjugates (compounds) and compositions, as well as methods of using them. The compounds of the invention are broad-spectrum, long-lasting, and non-toxic combination of synthetic immunostimulatory agents, which are useful for activating the immune system of a mammal, preferably a human and can help direct the pharmacophore to the receptor within the endosomes of target cells and enhance the signal transduction induced by the pharmacophore.

Description

RELATED APPLICATION
This application is a continuation of U.S. patent application Ser. No. 12/064,529, filed Feb. 5, 2009, which is a U.S. National Stage Filing under 35 U.S.C. 371 from International Application Number PCT/US2006/032371, filed Aug. 21, 2006 and published in English as WO 2007/024707 on Mar. 1, 2007, which claims the benefit of priority from U.S. Provisional Application Ser. No. 60/710,337 filed Aug. 22, 2005, under 35 U.S.C. 119(e), which applications and publication are herein incorporated by reference in their entirety.
GOVERNMENT FUNDING
The invention was made with government support under Grant Number AI57436 awarded by the National Institutes of Health. The government has certain rights in the invention.
BACKGROUND OF THE INVENTION
A great deal has been learned about the molecular basis of innate recognition of microbial pathogens in the last decade. It is generally accepted that many somatic cells express a range of pattern recognition receptors that detect potential pathogens independently of the adaptive immune system. (See Janeway et al., Annu Rev Immunol, 20:197-216 (2002).) These receptors are believed to interact with microbial components termed pathogen associated molecular patterns (PAMPs). Examples of PAMPs include peptidoglycans, lipotechoic acids from gram-positive cell walls, the sugar mannose (which is common in microbial carbohydrates but rare in humans), bacterial DNA, double-stranded RNA from viruses, and glucans from fungal cell walls. PAMPs generally meet certain criteria that include, (a) their expression by microbes but not their mammalian hosts, (b) conservation of structure across the wide range of pathogens, and (c) the capacity to stimulate innate immunity. Toll-like Receptors (TLRs) have been found to play a central role in the detection of PAMPs and in the early response to microbial infections. (See Underhill et al., Curr Opin Immunol, 14:103-110 (2002).) Ten mammalian TLRs and a number of their agonists have been identified. For example, TLR7 and TLR9 recognize and respond to imiquimod and immunostimulatory CpG oligonucleotides (ISS-ODN), respectively. The synthetic immunomodulator R-848 (resiquimod) activates both TLR7 and TLR8. While TLR stimulation initiates a common signaling cascade (involving the adaptor protein MyD88, the transcription factor NF-kB, and pro-inflammatory and effector cytokines), certain cell types tend to produce certain TLRs. For example, TLR7 and TLR9 are found predominantly on the internal faces of endosomes in dendritic cells (DCs) and B lymphocytes (in humans; mouse macrophages express TLR7 and TLR9). TLR8, on the other hand, is found in human blood monocytes. (See Hornung et al., J Immunol, 168:4531-4537 (2002)).
Interferons (INFs) are also involved in the efficient induction of an immune response, especially after viral infection (Brassard et al., J. Leukoc Biol, 71:568-581 (2002).) However, many viruses produce a variety of proteins that block interferon production or action at various levels. Antagonism of interferon is believed to be part of a general strategy to evade innate, as well as adaptive immunity. (See Levy et al., Cytokine Growth Factor Rev, 12:143-156 (2001).) While TLR agonists (TLR-L) may be sufficiently active for some methods of treatment, in some instances the microbial interferon antagonists could mitigate the adjuvant effects of synthetic TLR-L.
Accordingly, there exists a need for compounds that augment TLR-induced signal transduction, i.e., compounds that hinder viral or bacterial obstruction of interferon production or have the ability to modulate the innate immune system using the TLR agonists.
SUMMARY OF THE INVENTION
The present invention provides for TLR agonist conjugates (compounds) and compositions, as well as methods of using them. The compounds of the invention are broad-spectrum, long-lasting, and non-toxic combination of synthetic immunostimulatory agents, which are useful for activating the immune system of a mammal, preferably a human and can help direct the pharmacophore to the receptor within the endosomes of target cells and enhance the signal transduction induced by the pharmacophore. The compounds of the invention include a pharmacophore covalently bound to an auxiliary group. Accordingly there is provided a compound of the invention which is a compound of formula (I):
Figure US09359360-20160607-C00001
wherein X1 is —O—, —S—, or —NRc—;
wherein Rc hydrogen, C1-10alkyl, or C1-10alkyl substituted by C3-6-cycloalkyl, or Rc and R1 taken together with the nitrogen atom can form a heterocyclic ring or a substituted heterocyclic ring, wherein the substituents are hydroxy, C1-6alkyl, hydroxyC1-6alkylene, C1-6alkoxy, C1-6alkoxyC1-6alkylene, or cyano;
R1 is (C1-C10)alkyl, substituted (C1-C10)alkyl, C6-10aryl, or substituted C6-10aryl, C5-9heterocyclic, substituted C5-9heterocyclic;
each R2 is independently hydrogen, —OH, (C1-C6)alkyl, substituted (C1-C6)alkyl, (C1-C6)alkoxy, substituted (C1-C6)alkoxy, —C(O)—(C1-C6)alkyl (alkanoyl), substituted —C(O)—(C1-C6)alkyl, —C(O)—(C6-C10)aryl (aroyl), substituted —C(O)—(C6-C10)aryl, —C(O)OH (carboxyl), —C(O)O(C1-C6)alkyl (alkoxycarbonyl), substituted —C(O)O(C1-C6)alkyl, —NRaRb, —C(O)NRaRb (carbamoyl), substituted —C(O)NRaRb, halo, nitro, or cyano;
each Ra and Rb is independently hydrogen, (C1-C6)alkyl, (C3-C8)cycloalkyl, (C1-C6)alkoxy, halo(C1-C6)alkyl, (C3-C8)cycloalkyl(C1-C6)alkyl, (C1-C6)alkanoyl, hydroxy(C1-C6)alkyl, aryl, aryl(C1-C6)alkyl, Het, Het (C1-C6)alkyl, or (C1-C6)alkoxycarbonyl;
X2 is a bond or a linking group; and R3 is an auxiliary group;
n is 1, 2, 3, or 4; m is 1 or 2; q is 1 or 2; or
a pharmaceutically acceptable salt thereof.
The auxiliary groups can include organic molecules, composed of carbon, oxygen, hydrogen, nitrogen, sulfur, phosphorus atoms. These groups are not harmful to body tissues (e.g., they are non-toxic, and/or do not cause inflammation).
In addition, the invention also provides a pharmaceutical composition comprising at least one compound of formula (I), or a pharmaceutically acceptable salt thereof, in combination with a pharmaceutically acceptable diluent or carrier.
In one embodiment, the invention provides a therapeutic method for preventing or treating a pathological condition or symptom in a mammal, such as a human, wherein the activity of TLR agonists is implicated and its action is desired, comprising administering to a mammal in need of such therapy, an effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof. Non-limiting examples of pathological conditions or symptoms that are suitable for treatment include cancers, treatment for bacterial or viral diseases, treating autoimmune diseases, and treating Crohn's Disease.
The compounds of the invention can also be used as or to prepare vaccines against bacteria, viruses, cancer cells, cancer specific peptides, enhancers of monoclonal antibodies against cancer, a CNS stimulant, or for biodefense.
The invention provides a compound of formula (I) for use in medical therapy (e.g., for use as an anti-cancer agent, treatment for bacterial diseases, treatment for viral diseases, such as hepatitis C and hepatitis B, Crohn's Disease, and as therapeutic agents for treating immunologic disease). Furthermore, it is suggested that compounds of formula (I) will prevent carcinogenesis by hepatitis C and hepatitis B, as well as the use of a compound of formula (I) for the manufacture of a medicament useful for the treatment of cancer, viral diseases, Crohn's Disease, and immunologic disorders in a mammal, such as a human.
In a specific embodiment, the present invention provides a method for treating a viral infection in a mammal by administering a TLR agonist compound of formula (I). The viral infection can be caused by an RNA virus, a product of the RNA virus that acts as a TLR agonist and/or a DNA virus. A specific DNA virus for treatment is the Hepatitis B virus.
In another specific embodiment, the present invention provides a method for treating cancer by administering an effective amount of a TLR agonist compound of formula (I). The cancer can be an interferon sensitive cancer, such as, for example, a leukemia, a lymphoma, a myeloma, a melanoma, or a renal cancer.
In another specific embodiment, the present invention provides a method of treating an autoimmune disease by administering a therapeutically effective amount of a TLR agonist compound of formula (I) or a pharmaceutically acceptable salt of such a compound. A specific autoimmune disease is Multiple Sclerosis, lupus, rheumatoid arthritis and the like.
In another specific embodiment, the present invention provides a method of treating Crohn's Disease by administering a TLR agonist compound of formula (I).
The TLR agonists can be a homofunctional TLR agonist polymer and can consist of a TLR-7 agonist or a TLR-8 agonist. The TLR7 agonist can be a 7-thia-8-oxoguanosinyl (TOG) moiety, a 7-deazaguanosinyl (7DG) moiety, a resiquimod moiety, or an imiquimod moiety. The TLR8 agonist can be a resiquimod moiety. In another aspect, the TLR agonist is a heterofunctional TLR agonist polymer. The heterofunctional TLR agonist polymer can include a TLR-7 agonist and a TLR-8 agonist or a TLR-9 agonist or all three agonists. The heterofunctional TLR agonist polymer can include a TLR-8 agonist and a TLR-9 agonist.
The invention also provides processes and intermediates disclosed herein that are useful for preparing compounds of formula (I) or salts thereof.
BRIEF DESCRIPTION OF THE FIGURES
FIG. 1 is a graphic illustration of the absorption chromophore (at ˜350 nm) of a compound of formula I (OVA/IV150 Conjugate).
FIG. 2 is a graphic illustration of the stimulation of bone marrow derived dendritic cells (BMDC).
FIG. 3 illustrates the conjugation of a TLR7 agonist, UC-1V150, to mouse serum albumin (MSA). The success of conjugation is indicated by UV spectroscopy. The UC-1V150 to MSA ratio is approximately 5:1
FIGS. 4A and B illustrate that the UC-1V150 and MSA conjugates activate both murine bone marrow-derived macrophages (4A) and human peripheral blood mononuclear cells (4B). Cells were incubated with various concentrations of the compound from 0.5 nM to 10 μM in BMDM or from 0.1 to 10 μM in PBMC. Culture supernatants were harvested after 24 h and cytokine levels were analyzed by Luminex.
FIGS. 5A, 5B, 5C, and 5D illustrate the increased potency and duration of effect of UC-1V150/MSA. C57BL/6 mice were injected (i.v.) with (A) 0.1 micromole of SM-360320, a TLR7 ligand, or (B) equivalent amount of a TLR7 agonist UC-1V150 (aldehyde-modified SM-360320) or UC-1V150/MSA to 500 μg MSA per mouse. Serum samples were collected at the indicated time points and cytokine levels were analyzed by Luminex. MSA=mouse serum albumin. The effect from the original TLR7 ligand, SM-360320, lasted for only 2 hours whereas UC-1V150/MSA has extended the effect to at least 6 hours.
FIG. 6 illustrates the effects of UC-1V150 conjugated with inactivated SIV (6A) or with OVA in combination with ODN (6B). Myeloid BMDC were incubated for 24 hr with various conditions at 0.1 μg/ml as indicated. IL-12 levels in the cell supernatant were measured by ELISA.
FIGS. 7A and 7B illustrates an increased potency of UC-1V150/MSA. C57BL/6 mice were i.v. injected with 380 nmole of SM-360320 or UC-1V150, or 500 μg of UC-1V150/MSA (equivalent to 3.8 nmole UC-1V150) per mouse. Serum samples were collected after 2 h and cytokine levels were analyzed by Luminex. To achieve the similar effect, at least 100-fold higher concentration of either UC-1V150 or SM-360320 was required as compared to that of UC-1V150/MSA.
FIG. 8 is an illustration of the uv spectrum of a double-conjugate, (OVA/IV150/1043).
FIG. 9 is an illustration of the induction of IL-12 in BMDC using OVA/ODN/IV150 conjugates.
FIG. 10 illustrates direct conjugation of SIV Particles to the IA compound IV150.
FIG. 11 illustrates the ability to prepare compounds of the invention with virus particles attached to a compound having formula IA and the TLR agonist activity of the compounds.
FIG. 12 illustrates the molecular areas of specificity for antibodies raised against the conjugates containing a linker and a TLR ligand.
FIGS. 13A and 13B illustrate the distinction between the four substances applied to the respective lanes on a gel in a Western plot analysis. In FIG. 13A, the gel membrane was probed with anti-ovalbumin (anti-OVA) antibody and all lanes gave a positive band, indicating that OVA was detected in all lanes, as expected. In FIG. 13B, the gel membrane was probed with the selective antibody raised to the TLR ligand portion of the conjugate, and therefore only lane 4 was positive, confirming the specificity of the antibody for the TLR ligand.
DETAILED DESCRIPTION
Non-limiting examples of auxiliary groups include side chains that increase solubility, such as, for example, groups containing morpholino, piperidino, pyrrolidino, or piperazino rings and the like; amino acids, polymers of amino acids (proteins or peptides), e.g., dipeptides or tripeptides, and the like; carbohydrates (polysaccharides), nucleotides such as, for example, PNA, RNA and DNA, and the like; polymers of organic materials, such as, for example, polyethylene glycol, poly-lactide and the like; monomeric and polymeric lipids; insoluble organic nanoparticles; non-toxic body substances such as, for example, cells, lipids, vitamins, co-factors, antigens such as, for example microbes, such as, for example, viruses, bacteria, fungi, and the like. The antigens can include inactivated whole organisms, or sub-components thereof and the like.
The compounds of the invention can be prepared using compounds having formula (IA):
Figure US09359360-20160607-C00002

where X is a group that can react to form a bond to the linking group or can react to form a bond to the auxiliary group. A specific group of compounds having formula (IA) are disclosed in U.S. Pat. No. 6,329,381.
The following definitions are used, unless otherwise described: halo is fluoro, chloro, bromo, or iodo. Alkyl, alkoxy, alkenyl, alkynyl, etc. denote both straight and branched groups; but reference to an individual radical such as “propyl” embraces only the straight chain radical, a branched chain isomer such as “isopropyl” being specifically referred to. Aryl denotes a phenyl radical or an ortho-fused bicyclic carbocyclic radical having about nine to ten ring atoms in which at least one ring is aromatic. Heteroaryl encompasses a radical attached via a ring carbon of a monocyclic aromatic ring containing five or six ring atoms consisting of carbon and one to four heteroatoms each selected from the group consisting of non-peroxide oxygen, sulfur, and N(X) wherein X is absent or is H, O, (C1-C4)alkyl, phenyl or benzyl, as well as a radical of an ortho-fused bicyclic heterocycle of about eight to ten ring atoms derived therefrom, particularly a benz-derivative or one derived by fusing a propylene, trimethylene, or tetramethylene diradical thereto.
It will be appreciated by those skilled in the art that compounds of the invention having a chiral center may exist in and be isolated in optically active and racemic forms. Some compounds may exhibit polymorphism. It is to be understood that the present invention encompasses any racemic, optically-active, polymorphic, or stereoisomeric form, or mixtures thereof, of a compound of the invention, which possess the useful properties described herein, it being well known in the art how to prepare optically active forms (for example, by resolution of the racemic form by recrystallization techniques, by synthesis from optically-active starting materials, by chiral synthesis, or by chromatographic separation using a chiral stationary phase) and how to determine nicotine agonist activity using the standard tests described herein, or using other similar tests which are well known in the art.
Processes for preparing compounds of formula I or for preparing intermediates useful for preparing compounds of formula I are provided as further embodiments of the invention. Intermediates useful for preparing compounds of formula I are also provided as further embodiments of the invention.
In cases where compounds are sufficiently basic or acidic to form acid or base salts, use of the compounds as salts may be appropriate. Examples of acceptable salts are organic acid addition salts formed with acids which form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, α-ketoglutarate, and α-glycerophosphate. Suitable inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts.
Acceptable salts may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion. Alkali metal (for example, sodium, potassium or lithium) or alkaline earth metal (for example calcium) salts of carboxylic acids can also be made.
Alkyl includes straight or branched C1-10 alkyl groups, e.g., methyl, ethyl, propyl, butyl, pentyl, isopropyl, isobutyl, 1-methylpropyl, 3-methylbutyl, hexyl, and the like.
Lower alkyl includes straight or branched C1-6 alkyl groups, e.g., methyl, ethyl, propyl, 1-methylethyl, butyl, 1-methylpropyl, 2-methylpropyl, 1,1-dimethylethyl, pentyl, 1-methylbutyl, 2-methylbutyl, 3-methylbutyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, and the like.
The term “alkylene” refers to a divalent straight or branched hydrocarbon chain (e.g. ethylene —CH2—CH2—).
C3-7 Cycloalkyl includes groups such as, cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl, and the like, and alkyl-substituted C3-7 cycloalkyl group, preferably straight or branched C1-6 alkyl group such as methyl, ethyl, propyl, butyl or pentyl, and C5-7 cycloalkyl group such as, cyclopentyl or cyclohexyl, and the like.
Lower alkoxy includes C1-6 alkoxy groups, such as methoxy, ethoxy or propoxy, and the like.
Lower alkanoyl includes C1-6 alkanoyl groups, such as formyl, acetyl, propanoyl, butanoyl, pentanoyl or hexanoyl, and the like.
C7-11 aroyl, includes groups such as benzoyl or naphthoyl;
Lower alkoxycarbonyl includes C2-7 alkoxycarbonyl groups, such as methoxycarbonyl, ethoxycarbonyl or propoxycarbonyl, and the like.
Lower alkylamino group means amino group substituted by C1-6 alkyl group, such as, methylamino, ethylamino, propylamino, butylamino, and the like.
Di(lower alkyl)amino group means amino group substituted by the same or different and C1-6 alkyl group (e.g. dimethylamino, diethylamino, ethylmethylamino).
Lower alkylcarbamoyl group means carbamoyl group substituted by C1-6 alkyl group (e.g. methylcarbamoyl, ethylcarbamoyl, propylcarbamoyl, butylcarbamoyl).
Di(lower alkyl)carbamoyl group means carbamoyl group substituted by the same or different and C1-6 alkyl group (e.g. dimethylcarbamoyl, diethylcarbamoyl, ethylmethylcarbamoyl).
Halogen atom means halogen atom such as fluorine atom, chlorine atom, bromine atom or iodine atom.
Aryl refers to a C6-10 monocyclic or fused cyclic aryl group, such as phenyl, indenyl, or naphthyl, and the like.
Heterocyclic refers to monocyclic saturated heterocyclic groups, or unsaturated monocyclic or fused heterocyclic group containing at least one heteroatom, e.g., 0-3 nitrogen atoms (—NRd—), 0-1 oxygen atom (—O—), and 0-1 sulfur atom (—S—). Non-limiting examples of saturated monocyclic heterocyclic group includes 5 or 6 membered saturated heterocyclic group, such as tetrahydrofuranyl, pyrrolidinyl, morpholinyl, piperidyl, piperazinyl or pyrazolidinyl. Non-limiting examples of unsaturated monocyclic heterocyclic group includes 5 or 6 membered unsaturated heterocyclic group, such as furyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, pyridyl or pyrimidinyl. Non-limiting examples of unsaturated fused heterocyclic groups includes unsaturated bicyclic heterocyclic group, such as indolyl, isoindolyl, quinolyl, benzothizolyl, chromanyl, benzofuranyl, and the like.
Rc and R1 taken together with the nitrogen atom to which they are attached can form a heterocyclic ring. Non-limiting examples of heterocyclic rings include 5 or 6 membered saturated heterocyclic rings, such as 1-pyrrolidinyl, 4-morpholinyl, 1-piperidyl, 1-piperazinyl or 1-pyrazolidinyl, 5 or 6 membered unsaturated heterocyclic rings such as 1-imidazolyl, and the like.
The alkyl, aryl, heterocyclic groups of R1 can be optionally substituted with one or more substituents, wherein the substituents are the same or different, and include lower alkyl; cycloalkyl, hydroxyl; hydroxy C1-6 alkylene, such as hydroxymethyl, 2-hydroxyethyl or 3-hydroxypropyl; lower alkoxy; C1-6 alkoxy C1-6 alkyl, such as 2-methoxyethyl, 2-ethoxyethyl or 3-methoxypropyl; amino; alkylamino; dialkyl amino; cyano; nitro; acyl; carboxyl; lower alkoxycarbonyl; halogen; mercapto; C1-6 alkylthio, such as, methylthio, ethylthio, propylthio or butylthio; substituted C1-6 alkylthio, such as methoxyethylthio, methylthioethylthio, hydroxyethylthio or chloroethylthio; aryl; substituted C6-10 monocyclic or fused-cyclic aryl, such as 4-hydroxyphenyl, 4-methoxyphenyl, 4-fluorophenyl, 4-chlorophenyl or 3,4-dichlorophenyl; 5-6 membered unsaturated heterocyclic, such as furyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, thienyl, pyridyl or pyrimidinyl; and bicyclic unsaturated heterocyclic, such as indolyl, isoindolyl, quinolyl, benzothiazolyl, chromanyl, benzofuranyl or phthalimino.
The alkyl, aryl, heterocyclic groups of R2 can be optionally substituted with one or more substituents, wherein the substituents are the same or different, and include hydroxyl; C1-6 alkoxy, such as methoxy, ethoxy or propoxy; carboxyl; C2-7 alkoxycarbonyl, such as methoxycarbonyl, ethoxycarbonyl or propoxycarbonyl) and halogen.
The alkyl, aryl, heterocyclic groups of Rc can be optionally substituted with one or more substituents, wherein the substituents are the same or different, and include C3-6 cycloalkyl; hydroxyl; C1-6 alkoxy; amino; cyano; aryl; substituted aryl, such as 4-hydroxyphenyl, 4-methoxyphenyl, 4-chlorophenyl or 3,4-dichlorophenyl; nitro and halogen.
The heterocyclic ring formed together with Rc and R1 and the nitrogen atom to which they are attached can be optionally substituted with one or more substituents, wherein the substituents are the same or different, and include C1-6 alkyl; hydroxy C1-6 alkylene; C1-6 alkoxy C1-6 alkylene; hydroxyl; C1-6 alkoxy; and cyano.
The term “amino acid” as used herein, comprises the residues of the natural amino acids (e.g. Ala, Arg, Asn, Asp, Cys, Glu, Gln, Gly, His, Hyl, Hyp, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, and Val) in D or L form, as well as unnatural amino acids (e.g. phosphoserine, phosphothreonine, phosphotyrosine, hydroxyproline, gamma-carboxyglutamate; hippuric acid, octahydroindole-2-carboxylic acid, statine, 1,2,3,4,-tetrahydroisoquinoline-3-carboxylic acid, penicillamine, ornithine, citruline, -methyl-alanine, para-benzoylphenylalanine, phenylglycine, propargylglycine, sarcosine, and tert-butylglycine). The term also comprises natural and unnatural amino acids bearing a conventional amino protecting group (e.g. acetyl or benzyloxycarbonyl), as well as natural and unnatural amino acids protected at the carboxy terminus (e.g. as a (C1-C6)alkyl, phenyl or benzyl ester or amide; or as an -methylbenzyl amide). Other suitable amino and carboxy protecting groups are known to those skilled in the art (See for example, T. W. Greene, Protecting Groups In Organic Synthesis; Wiley: New York, 1981, and references cited therein). An amino acid can be linked to the remainder of a compound of formula I through the carboxy terminus, the amino terminus, or through any other convenient point of attachment, such as, for example, through the sulfur of cysteine.
The term “toll-like receptor” (TLR) refers to a member of a family of receptors that bind to pathogen associated molecular patterns (PAMPs) and facilitate an immune response in a mammal. Ten mammalian TLRs are known, e.g., TLR1-10.
The term “toll-like receptor agonist” (TLR agonist) refers to a molecule that binds to a TLR and antagonizes the receptor. Synthetic TLR agonists are chemical compounds that are designed to bind to a TLR and activate the receptor. Exemplary novel TLR agonists provided herein include “TLR-7 agonist” “TLR-8 agonist” and “TLR-9 agonist.”
The term “nucleic acid” as used herein, refers to DNA, RNA, single-stranded, double-stranded, or more highly aggregated hybridization motifs, and any chemical modifications thereof. Modifications include, but are not limited to, those providing chemical groups that incorporate additional charge, polarizability, hydrogen bonding, electrostatic interaction, and fluxionality to the nucleic acid ligand bases or to the nucleic acid ligand as a whole. Such modifications include, but are not limited to, peptide nucleic acids (PNAs), phosphodiester group modifications (e.g., phosphorothioates, methylphosphonates), 2′-position sugar modifications, 5-position pyrimidine modifications, 7-position purine modifications, 8-position purine modifications, 9-position purine modifications, modifications at exocyclic amines, substitution of 4-thiouridine, substitution of 5-bromo or 5-iodo-uracil; backbone modifications, methylations, unusual base-pairing combinations such as the isobases, isocytidine and isoguanidine and the like. Nucleic acids can also include non-natural bases, such as, for example, nitroindole. Modifications can also include 3′ and 5′ modifications such as capping with a BHQ, a fluorophore or another moiety.
A specific value for X1 is a sulfur atom, an oxygen atom or —NRc—.
Another specific X1 is a sulfur atom.
Another specific X1 is an oxygen atom.
Another specific X1 is —NRc—.
Another specific X1 is —NH—.
A specific value for Rc is hydrogen, C1-4 alkyl or substituted C1-4 alkyl.
A specific value for R1 and Rc taken together is when they form a heterocyclic ring or a substituted heterocyclic ring.
Another specific value for R1 and Rc taken together is substituted or unsubstituted morpholino, piperidino, pyrrolidino, or piperazino ring.
A specific value for R1 is hydrogen, C1-4alkyl, or substituted C1-4alkyl.
Another specific R1 is 2-hydroxyethyl, 3-hydroxypropyl, 4-hydroxybutyl, 2-aminoethyl, 3-aminopropyl, 4-aminobutyl, methoxymethyl, 2-methoxyethyl, 3-methoxypropyl, ethoxymethyl, 2-ethoxyethyl, methylthiomethyl, 2-methylthioethyl, 3-methylthiopropyl, 2-fluoroethyl, 3-fluoropropyl, 2,2,2-trifluoroethyl, cyanomethyl, 2-cyanoethyl, 3-cyanopropyl, methoxycarbonylmethyl, 2-methoxycarbonylethyl, 3-methoxycarbonylpropyl, benzyl, phenethyl, 4-pyridylmethyl, cyclohexylmethyl, 2-thienylmethyl, 4-methoxyphenylmethyl, 4-hydroxyphenylmethyl, 4-fluorophenylmethyl, or 4-chlorophenylmethyl.
Another specific R1 is hydrogen, CH3—, CH3—CH2—, CH3CH2CH2—, hydroxyC1-4alkylene, or C1-4alkoxyC1-4alkylene.
Another specific value for R1 is hydrogen, CH3—, CH3—CH2—, CH3—O—CH2CH2— or CH3—CH2—O—CH2CH2—.
A specific value for R2 is hydrogen, halogen, or C1-4alkyl.
Another specific value for R2 is hydrogen, chloro, bromo, CH3—, or CH3—CH2—.
Specific substituents for substitution on the alkyl, aryl or heterocyclic groups are hydroxy, C1-6alkyl, hydroxyC1-6alkylene, C1-6alkoxy, C1-6alkoxyC1-6alkylene, C3-6cycloalkyl, amino, cyano, halogen, or aryl.
A specific value for X2 is a bond or a chain having up to about 24 atoms; wherein the atoms are selected from the group consisting of carbon, nitrogen, sulfur, non-peroxide oxygen, and phosphorus.
Another specific value for X2 is a bond or a chain having from about 4 to about 12 atoms.
Another specific value for X2 is a bond or a chain having from about 6 to about 9 atoms.
Another specific value for X2 is
Figure US09359360-20160607-C00003
Another specific value for X2 is
Figure US09359360-20160607-C00004
A specific auxiliary group is an amino acid, a carbohydrate, a peptide, an antigen, a nucleic acid, a body substance, or a microbe.
A specific peptide, has from 2 to about 20 amino acid residues.
Another specific peptide, has from 10 to about 20 amino acid residues.
A specific auxiliary group is a carbohydrate.
A specific nucleic acid is DNA, RNA or PNA.
A specific body substance is a cell, lipid, vitamin, or co-factor.
Another specific body substance is a cell or lipid.
A specific antigen is a microbe.
A specific microbe is a virus, bacteria, or fungi.
Another specific microbe is a virus or a bacteria.
Specific bacteria are Bacillus anthracis (anthrax), Listeria monocytogenes, Francisella tularensis, or Salmonella.
Specific Salmonella are typhimurium or enteritidis.
Specific viruses are RNA viruses, a product of the RNA virus, or a DNA virus.
A specific DNA virus is the Hepatitis B virus.
Specific compounds of the invention have the general formula
IA-L-A1;
IA-L-(A1)2;
IA-L-A1-A1;
IA-L-A1-L-A1;
(IA)2-L-A1-A1;
(IA)2-L-A1-L-A1;
(IA)2-L-A1; or
(IA)2-L-(A1)2;
wherein IA is as disclosed herein; L is absent or is a linking group; and each A1 group independently represents an auxiliary group.
In one embodiment, the viral infection is caused by a coronavirus that causes Severe Acute Respiratory Syndrome (SARS), a Hepatitis B virus, or a Hepatitis C Virus.
In another embodiment, the viral infection is caused by a coronavirus that causes Severe Acute Respiratory Syndrome (SARS), a Hepatitis B virus, or a Hepatitis C Virus.
Specific cancers that can be treated include melanoma, superficial bladder cancer, actinic keratoses, intraepithelial neoplasia, and basal cell skin carcinoma, squamous, and the like. In addition, the method of the invention includes treatment for a precancerous condition such as, for example, actinic keratoses or intraepithelial neoplasia, familial polyposis (polyps), cervical dysplasia, cervical cancers, superficial bladder cancer, and any other cancers associated with infection (e.g., lymphoma Karposi's sarcoma, or leukemia); and the like.
Non limiting examples of the pathological conditions or symptoms that can be treated include viral diseases, cancer, inflammatory diseases of the gastrointestinal tract, brain, skin, joints, and other tissues.
The auxiliary groups are believed to enhance the drug activity of the pharmacophore (compounds of formula (I)) by (a) helping to direct the pharmacophore to the receptor within the endosomes of target cells; (b) by enhancing signal transduction induced by the pharmacophore, by cross-linking the receptor; and/or (c) the pharmacophore can enhance the response to the auxiliary group (e.g., immune response). The auxiliary groups should form generally stable bonds with the pharmacophore, and do not act as prodrugs.
The invention includes compositions of a compound of formula (I) optionally in combination with an inhibitor of inosine monophosphate dehydrogenase (IMPDH), an enantiomer of such a compound, a prodrug of such a compound, or a pharmaceutically acceptable salt of such a compound. As used herein an “IMPDH inhibitor” refers to an inhibitor of the enzyme inosine monophosphate dehydrogenase. Currently, three IMPDH inhibitors are used clinically: ribavirin, mizoribine, and mycophenolate mofetil. Ribavirin and mizoribine are prodrugs that are phosphorylated intracellularly to produce IMP analogs (Goldstein et al., Cuff Med Chem, 6:519-536 (1999)). Viramidine is a prodrug of Ribavirin. Mycophenolate mofetil is immunosuppressive, and has gastrointestinal irritative properties that may be attributable to its enterohepatic recirculation (Papageorgiou C, Mini Rev Med Chem., 1:71-77 (2001)). Mizoribine aglycone, a prodrug, is used as an IMPDH inhibitor. Other non-limiting examples IMPDH inhibitors, including prodrugs of mizoribine and mizoribine aglycone are known and are disclosed in published U.S. Patent application No. 20050004144.
In cases where compounds are sufficiently basic or acidic to form stable nontoxic acid or base salts, administration of the compounds as salts may be appropriate. Examples of pharmaceutically acceptable salts are organic acid addition salts formed with acids which form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartarate, succinate, benzoate, ascorbate, α-ketoglutarate, and α-glycerophosphate. Suitable inorganic salts may also be formed, including hydrochloride, sulfate, nitrate, bicarbonate, and carbonate salts.
Pharmaceutically acceptable salts may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion. Alkali metal (for example, sodium, potassium or lithium) or alkaline earth metal (for example calcium) salts of carboxylic acids can also be made.
The compounds (conjugates) of the invention can be prepared using standard synthetic methods known in the art. A general ester synthesis is illustrated below:
Figure US09359360-20160607-C00005
Figure US09359360-20160607-C00006
Additional examples for preparing specific compounds are included herein.
The compounds of formula I can be formulated as pharmaceutical compositions and administered to a mammalian host, such as a human patient in a variety of forms adapted to the chosen route of administration, i.e., orally or parenterally, by intravenous, intramuscular, topical or subcutaneous routes.
Thus, the present compounds may be systemically administered, e.g., orally, in combination with a pharmaceutically acceptable vehicle such as an inert diluent or an assimilable edible carrier. They may be enclosed in hard or soft shell gelatin capsules, may be compressed into tablets, or may be incorporated directly with the food of the patient's diet. For oral therapeutic administration, the active compound may be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Such compositions and preparations should contain at least 0.1% of active compound. The percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 60% of the weight of a given unit dosage form. The amount of active compound in such therapeutically useful compositions is such that an effective dosage level will be obtained.
The tablets, troches, pills, capsules, and the like may also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring may be added. When the unit dosage form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier, such as a vegetable oil or a polyethylene glycol. Various other materials may be present as coatings or to otherwise modify the physical form of the solid unit dosage form. For instance, tablets, pills, or capsules may be coated with gelatin, wax, shellac or sugar and the like. A syrup or elixir may contain the active compound, sucrose or fructose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor. Of course, any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed. In addition, the active compound may be incorporated into sustained-release preparations and devices.
The active compound may also be administered intravenously or intraperitoneally by infusion or injection. Solutions of the active compound or its salts can be prepared in water, optionally mixed with a nontoxic surfactant. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
The pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes. In all cases, the ultimate dosage form should be sterile, fluid and stable under the conditions of manufacture and storage. The liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
Sterile injectable solutions are prepared by incorporating the active compound in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filter sterilization. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
For topical administration, the present compounds may be applied in pure form, i.e., when they are liquids. However, it will generally be desirable to administer them to the skin as compositions or formulations, in combination with a dermatologically acceptable carrier, which may be a solid or a liquid.
Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like. Useful liquid carriers include water, alcohols or glycols or water-alcohol/glycol blends, in which the present compounds can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants. Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use. The resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers.
Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.
Examples of useful dermatological compositions which can be used to deliver the compounds of formula I to the skin are known to the art; for example, see Jacquet et al. (U.S. Pat. No. 4,608,392), Geria (U.S. Pat. No. 4,992,478), Smith et al. (U.S. Pat. No. 4,559,157) and Wortzman (U.S. Pat. No. 4,820,508).
Useful dosages of the compounds of formula I can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949.
Generally, the concentration of the compound(s) of formula I in a liquid composition, such as a lotion, will be from about 0.1-25 wt-%, preferably from about 0.5-10 wt-%. The concentration in a semi-solid or solid composition such as a gel or a powder will be about 0.1-5 wt-%, preferably about 0.5-2.5 wt-%.
The amount of the compound, or an active salt or derivative thereof, required for use in treatment will vary not only with the particular salt selected but also with the route of administration, the nature of the condition being treated and the age and condition of the patient and will be ultimately at the discretion of the attendant physician or clinician.
In general, however, a suitable dose will be in the range of from about 0.5 to about 100 mg/kg, e.g., from about 10 to about 75 mg/kg of body weight per day, such as 3 to about 50 mg per kilogram body weight of the recipient per day, preferably in the range of 6 to 90 mg/kg/day, most preferably in the range of 15 to 60 mg/kg/day.
The compound is conveniently administered in unit dosage form; for example, containing 5 to 1000 mg, conveniently 10 to 750 mg, most conveniently, 50 to 500 mg of active ingredient per unit dosage form.
Ideally, the active ingredient should be administered to achieve peak plasma concentrations of the active compound of from about 0.5 to about 75 μM, preferably, about 1 to 50 μM, most preferably, about 2 to about 30 μM. This may be achieved, for example, by the intravenous injection of a 0.05 to 5% solution of the active ingredient, optionally in saline, or orally administered as a bolus containing about 1-100 mg of the active ingredient. Desirable blood levels may be maintained by continuous infusion to provide about 0.01-5.0 mg/kg/hr or by intermittent infusions containing about 0.4-15 mg/kg of the active ingredient(s).
The desired dose may conveniently be presented in a single dose or as divided doses administered at appropriate intervals, for example, as two, three, four or more sub-doses per day. The sub-dose itself may be further divided, e.g., into a number of discrete loosely spaced administrations; such as multiple inhalations from an insufflator or by application of a plurality of drops into the eye.
The ability of a compound of the invention to act as a TLR agonist may be determined using pharmacological models which are well known to the art, including the procedures disclosed by Lee et al.; PNAS, 100 p 6646-6651, 2003.
Processes for preparing compounds of formula (I) are provided as further embodiments of the invention and are illustrated by the following procedures in which the meanings of the generic radicals are as given above unless otherwise qualified.
EXAMPLES
General Chemistry.
Reagents and solvents were acquired from Aldrich, Milwaukee, Wis. Uncorrected melting points were determined on a Laboratory Device Mel-Temp II capillary melting point apparatus. Proton nuclear magnetic resonance spectra were recorded on a Varian Unity 500 NMR spectrophotometer at 499.8 MHz or on a Varian Mercury NMR spectrophotometer at 400.06 MHz. The chemical shifts were reported in ppm on the scale from the indicated reference. Positive and negative ion loop mass spectra were performed by Department of Chemistry UCSD, San Diego, Calif. Elemental analyses were performed by NuMega Resonance Labs, San Diego, Calif. Column chromatography was conducted on E Merck silica gel (230-400 mesh) with the indicated solvent system. Analytical thin layer chromatography (TLC) was conducted on silica gel 60 F-254 plates (EM Reagents).
Example 1 Preparation of 4-(2,6-dichloropurin-9-ylmethyl)benzonitrile
2,6-dichloro-9H-purine (16 mmol) is dissolved in DMF (50 mL) and potassium carbonate (50 mmol) is added. α-Bromo-p-tolunitrile (22 mmol) is then added and the mixture is stirred at ambient temperature for 16 h. After filtration to remove insoluble inorganic salts, the filtrate is poured into water (1500 mL) and extracted with ethyl acetate (2×400 mL), dried over magnesium sulfate and evaporated to yield a residue which is subjected to flash silica gel chromatography using 1:2:10 ethyl acetate/acetone/hexanes. Yield 3.33 g (69%). UV, NMR and MS were consistent with structure assignment.
Example 2 Preparation of 4-(6-amino-2-chloropurin-9-ylmethylbenzonitrile
The product of example 1 (1.9 g) is placed in a steel reaction vessel and methanolic ammonia (80 mL, 7 N) is added. The sealed vessel is heated at 60° C. for 12 h, cooled in ice and the solid product filtered off. Yield 1.09 g. UV, NMR and MS were consistent with assigned structure.
Example 3 Preparation of 4-[6-amino-2-(2-methoxyethoxy)purin-9-ylmethyl]benzonitrile
Sodium salt of 2-methoxyethanol is generated by dissolving sodium metal (81 mg) in 2-methoxyethanol (30 mL) with heat. To this solution is added the product of example 2 (1.0 g) dissolved in methoxyethanol (300 mL, with heat). The reaction mixture is heated for 8 h at 115° C. bath temperature, concentrated in vacuo to near dryness and the residue partitioned between ethyl acetate and water. Flash silica gel chromatography of the organic layer using 5% methanol in dichloromethane gave 763 mg product. NMR is consistent with structure assignment.
Example 4 Preparation of 4-[6-amino-8-bromo-2-(2-methoxyethoxy)purin-9-ylmethyl]benzonitrile
The product of example 3 (700 mg) is dissolved in dichloromethane (400 mL) and bromine (7 mL) is added dropwise. The mixture is stirred overnight at room temperature and extracted with aqueous sodium thiosulfate (2 L of 0.1 M) solution and then with aqueous sodium bicarbonate (500 mL, saturated). The residue from the organic layer is chromatographed on silica gel using 3% methanol in dichloromethane) to yield 460 mg of bromo product. NMR, UV and MS are consistent with structure assignment.
Example 5 Preparation of 4-[6-amino-8-methoxy-2-(2-methoxyethoxy)purin-9-ylmethyl]benzonitrile
Sodium methoxide is generated by reaction of sodium metal (81 mg) in dry methanol (30 mL). The product of example 4 (700 mg) is dissolved in dry dimethoxyethane and the temperature raised to 100° C. After overnight reaction, the mixture is concentrated in vacuo and the residue is chromatographed on silica using 5% methanol in dichloromethane. Yield 120 mg. NMR is consistent with structure assignment.
Example 6 Preparation of Lithium N,N′-(dimethylethylenediamino)aluminum hydride
This reducing agent used to convert the nitrile to the aldehyde function is prepared essentially as described in Bull. Korean Chem. Soc. (2002), 23(12), 1697-1698. A 0.5 M solution in dry THF is prepared.
Example 7 Preparation of 4-[6-amino-8-methoxy-2-(2-methoxyethoxy)purin-9-ylmethyl]benzaldehyde
The product of example 5 (100 mg) is dissolved in dry THF (3 mL) and cooled to 0° C. under argon. The reagent generated in example 6 (0.72 mL) is added to the reaction flask and the mixture is stirred at 0-5° C. for 1 h and then quenched by addition of 3 M HCl. The mixture is then extracted with ethyl acetate and then dichloromethane and concentrated in vacuo to yield 85 mg. NMR is consistent with structure assignment.
Example 8 Preparation of 4-[6-amino-8-hydroxy-2-(2-methoxyethoxy)purin-9-ylmethyl]benzaldehyde (1V150)
The product of example 7 (800 mg) is combined with sodium iodide (504 mg) and acetonitrile (40 mL), and then chlorotrimethylsilane (0.5 mL) is slowly added. The mixture is heated at 70° C. for 3.5 h, cooled and filtered. The solid product is washed with water, then ether to yield 406 mg. NMR, UV, MS are consistent with structure assignment. This material is suitable for conjugation reactions between linkers and auxiliary groups.
Example 9 Preparation of methyl 4-[6-amino-8-methoxy-2-(2-methoxyethoxy)purin-9-ylmethyl]benzoate. (Procedure as Described by Jayachitra, et al., Synth. Comm., (2003) 33(19), 3461-3466.)
The product of example 5 (1 mmol) is dissolved in dry methanol (5 mL) and freshly distilled BF3 etherate (4 mmol) is added to the solution. The resulting mixture is refluxed under argon for 20 h. The solvent is removed in vacuo and the residue is taken up in dichloromethane (10 mL) and extracted with dilute aqueous sodium bicarbonate (2×10 mL) and the organic layer is dried over magnesium sulfate. After evaporation the product is purified by silica gel column chromatography using 5% methanol in dichloromethane to yield 0.8 mmol.
Example 10 Preparation of 4-[6-amino-8-hydroxy-2-(2-methoxyethoxy)purin-9-ylmethyl]benzoic acid
The product of example 9 (100 mg) is combined with sodium iodide (63 mg) and acetonitrile (10 mL), and then chlorotrimethylsilane (120 mL) is slowly added. The mixture is heated at 70° C. for 6 h, cooled and filtered. The solid product is washed with water, then ether to yield 51 mg.
Example 11 Preparation of 2,5-dioxopyrrolidin-1-yl 4-[6-amino-8-hydroxy-2-(2-methoxyethoxy)purin-9-ylmethyl]benzoate
The product of example 10 (2 mmol) is dissolved in dichloromethane or dioxane (10 mL) and EDC (2 mmol) is added. To this solution is added N-hydroxysuccinimide (2 mmol) and resulting mixture is stirred at room temperature for 1 h. The mixture is taken to dryness in vacuo and the crude product is purified by silica gel chromatography to yield 2 mmol of product that is suitable for conjugation reactions involving primary amines.
Example 12 Conjugation of 1V150 to Mouse Serum Albumin (MSA)
Modification of MSA with SANH: 200 μl of MSA (25 mg/ml) was mixed with 100 μl of conjugation buffer (1M NaPi, pH=7.2) and 690 μl of PBS. 844 μg of SANH in 10 μl of DFM (40-fold molar excess to MSA) was added to protein solution (Final concentration of MSA in reaction mixture is 5 mg/ml). After gentle mixing reaction was proceeded at room temperature for 2 hr. To remove excess of SANH the reaction mixture was loaded on NAP-10 column equilibrated with PBS and modified MSA was eluted with 1.5 ml of PBS.
Attachment of IV150 to MSA modified with SANH: 460 μg of IV150 dissolved in 10 μl of DMF was added to MSA modified with SANH and the reaction mixture was incubated at RT overnight. To remove excess of IV150 the reaction mixture was firstly concentrated to 1 ml using micro-spin column (Millipore: BIOMAX 5K) and loaded on NAP-10 column as mentioned above.
The abbreviations used herein have their conventional meaning within the chemical and biological arts. All publications, patents, and patent documents cited in the specification are incorporated by reference herein, as though individually incorporated by reference. In the case of any inconsistencies, the present disclosure, including any definitions therein will prevail. The invention has been described with reference to various specific and preferred embodiments and techniques. However, it should be understood that many variations and modifications may be made while remaining within the spirit and scope of the invention.

Claims (13)

What is claimed is:
1. A compound having formula (IA) and R3:
Figure US09359360-20160607-C00007
wherein X1 is —O;
R1 is (C1-C10)alkyl or substituted (C1-C10)alkyl, wherein the substituents are hydroxy, C1-6alkyl, hydroxyC1-6alkylene, or C1-6alkoxy;
wherein n=4 and each R2 is —H;
X2 is a bond or a linking group having up to about 24 atoms; wherein the atoms in the chain of the linking group are selected from the group consisting of carbon, nitrogen, sulfur, non-peroxide oxygen, and phosphorous; and R3 is a protein that directs formula (IA) to receptors within endosomes of target cells;
or a pharmaceutically acceptable salt of the compound,
wherein the ratio of the compound having formula (IA) to R3 is 5:1.
2. The compound of claim 1, wherein R1 is C1-4alkyl, or substituted C1-4alkyl.
3. The compound of claim 2, wherein R1 is CH3—, CH3—C2—, CH3CH2CH2—, hydroxyC1-4alkylene, or C1-4alkoxyC1-4alkylene.
4. The compound of claim 3, wherein R1 is CH3—, CH3—CH2—, CH3—O—CH2CH2— or CH3—CH2—O—CH2CH2—.
5. The compound of claim 1, wherein X2 is a bond or a chain having from about 4 to about 12 atoms.
6. The compound of claim 1, wherein X2 is a bond or a chain having from about 6 to about 9 atoms.
7. The compound of claim 1, wherein X2 is
Figure US09359360-20160607-C00008
8. The compound of claim 1, wherein the protein is albumin.
9. The compound of claim 1, wherein the protein is ovalbumin.
10. A compound having formula (IA) and R3:
Figure US09359360-20160607-C00009
wherein X1 is —O—;
R1 is (C1-C10)alkyl or substituted (C1-C10)alkyl, wherein the substituents are hydroxy, C1-6alkyl, hydroxyC1-6alkylene, or C1-6alkoxy;
each R2 is —H and n=4;
X2 is a bond or a linking group having up to about 24 atoms; wherein the atoms in the chain of the linking group are selected from the group consisting of carbon, nitrogen, sulfur, and non-peroxide oxygen; and R3 is a protein that directs formula (IA) to receptors within endosomes of target cells;
or a pharmaceutically acceptable salt of the compound,
wherein the ratio of the compound having formula (IA) to R3 is 5:1.
11. A pharmaceutical composition comprising the compound of claim 10, and a pharmaceutically acceptable carrier.
12. The compound of claim 10, wherein the protein is albumin.
13. The compound of claim 10, wherein the protein is ovalbumin.
US13/682,208 2005-08-22 2012-11-20 TLR agonists Active US9359360B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/682,208 US9359360B2 (en) 2005-08-22 2012-11-20 TLR agonists

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US71033705P 2005-08-22 2005-08-22
PCT/US2006/032371 WO2007024707A2 (en) 2005-08-22 2006-08-21 Tlr agonists
US6452909A 2009-02-05 2009-02-05
US13/682,208 US9359360B2 (en) 2005-08-22 2012-11-20 TLR agonists

Related Parent Applications (3)

Application Number Title Priority Date Filing Date
US12/064,529 Continuation US20090324551A1 (en) 2005-08-22 2006-08-21 Tlr agonists
PCT/US2006/032371 Continuation WO2007024707A2 (en) 2005-08-22 2006-08-21 Tlr agonists
US6452909A Continuation 2005-08-22 2009-02-05

Publications (2)

Publication Number Publication Date
US20130165455A1 US20130165455A1 (en) 2013-06-27
US9359360B2 true US9359360B2 (en) 2016-06-07

Family

ID=37772216

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/064,529 Abandoned US20090324551A1 (en) 2005-08-22 2006-08-21 Tlr agonists
US13/682,208 Active US9359360B2 (en) 2005-08-22 2012-11-20 TLR agonists

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US12/064,529 Abandoned US20090324551A1 (en) 2005-08-22 2006-08-21 Tlr agonists

Country Status (7)

Country Link
US (2) US20090324551A1 (en)
EP (1) EP1931352B1 (en)
JP (2) JP2009504803A (en)
CN (1) CN101304748A (en)
CA (1) CA2620182A1 (en)
ES (1) ES2577514T3 (en)
WO (1) WO2007024707A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018112223A1 (en) 2016-12-14 2018-06-21 Progenity Inc. Treatment of a disease of the gastrointestinal tract with a tlr modulator
WO2021174024A1 (en) 2020-02-28 2021-09-02 First Wave Bio, Inc. Methods of treating iatrogenic autoimmune colitis
WO2022003598A1 (en) 2020-07-02 2022-01-06 Viiv Healthcare Company Method of achieving hiv viral remission using long-acting antiretroviral agents
US11697851B2 (en) 2016-05-24 2023-07-11 The Regents Of The University Of California Early ovarian cancer detection diagnostic test based on mRNA isoforms
EP4252629A2 (en) 2016-12-07 2023-10-04 Biora Therapeutics, Inc. Gastrointestinal tract detection methods, devices and systems

Families Citing this family (63)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101111085B1 (en) * 2002-09-27 2012-04-12 다이닛본 스미토모 세이야꾸 가부시끼가이샤 Novel adenine compound and use thereof
US8012964B2 (en) 2004-03-26 2011-09-06 Dainippon Sumitomo Pharma Co., Ltd. 9-substituted 8-oxoadenine compound
WO2007024707A2 (en) 2005-08-22 2007-03-01 The Regents Of The University Of California Tlr agonists
WO2007034817A1 (en) * 2005-09-22 2007-03-29 Dainippon Sumitomo Pharma Co., Ltd. Novel adenine compound
EP2029597A4 (en) 2006-05-31 2011-11-23 Univ California Purine analogs
EP2041135A4 (en) 2006-07-05 2010-12-01 Astrazeneca Ab 8-oxoadenine derivatives acting as modulators of tlr7
TW200831105A (en) * 2006-12-14 2008-08-01 Astrazeneca Ab Novel compounds
PL2125007T3 (en) 2007-02-07 2014-07-31 Univ California Conjugates of synthetic tlr agonists and uses therefor
PT2125792E (en) * 2007-02-19 2011-03-01 Glaxosmithkline Llc Purine derivatives as immunomodulators
SI2132209T1 (en) * 2007-03-19 2014-05-30 Astrazeneca Ab 9-substituted-8-oxo-adenine compounds as toll-like receptor (tlr7 ) modulators
US8063051B2 (en) * 2007-03-19 2011-11-22 Astrazeneca Ab 9-substituted-8-oxo-adenine compounds as toll-like receptor (TLR7) modulators
JPWO2008114819A1 (en) 2007-03-20 2010-07-08 大日本住友製薬株式会社 New adenine compounds
AR065784A1 (en) * 2007-03-20 2009-07-01 Dainippon Sumitomo Pharma Co DERIVATIVES OF 8-OXO ADENINE, DRUGS THAT CONTAIN THEM AND USES AS THERAPEUTIC AGENTS FOR ALLERGIC, ANTIVIRAL OR ANTIBACTERIAL DISEASES.
BRPI0811125A2 (en) * 2007-05-08 2017-05-09 Astrazeneca Ab imidazoquinolines with immunomodulatory properties
ES2541434T3 (en) 2007-06-29 2015-07-20 Gilead Sciences, Inc. Purine derivatives and their use as modulators of the Toll 7 receptor
WO2009034386A1 (en) * 2007-09-13 2009-03-19 Astrazeneca Ab Derivatives of adenine and 8-aza-adenine and uses thereof-796
PE20091156A1 (en) 2007-12-17 2009-09-03 Astrazeneca Ab SALTS OF (3 - {[[3- (6-AMINO-2-BUTOXY-8-OXO-7,8-DIHIDRO-9H-PURIN-9-IL) PROPYL] (3-MORFOLIN-4-ILPROPIL) AMINO] METHYL} PHENYL) METHYL ACETATE
CN102143964A (en) 2008-01-17 2011-08-03 大日本住友制药株式会社 Method for producing adenine compound
JPWO2009091031A1 (en) * 2008-01-17 2011-05-26 大日本住友製薬株式会社 Method for producing adenine compound
US20090202626A1 (en) * 2008-02-07 2009-08-13 Carson Dennis A Treatment of bladder diseases with a tlr7 activator
UA103195C2 (en) * 2008-08-11 2013-09-25 Глаксосмитклайн Ллк Purine derivatives for use in the treatment of allergic, inflammatory and infectious diseases
NZ593110A (en) 2008-12-09 2013-06-28 Gilead Sciences Inc pteridinone derivatives as MODULATORS OF TOLL-LIKE RECEPTORS
WO2010088395A2 (en) * 2009-01-30 2010-08-05 Idera Pharmaceuticals, Inc. Novel synthetic agonists of tlr9
WO2010088924A1 (en) 2009-02-06 2010-08-12 Telormedix Sa Pharmaceutical compositions comprising imidazoquinolin(amines) and derivatives thereof suitable for local administration
US8729088B2 (en) 2009-02-11 2014-05-20 The Regents Of The University Of California Toll-like receptor modulators and treatment of diseases
CN102666541B (en) 2009-10-22 2015-11-25 吉里德科学公司 Be used for the treatment of the particularly purine of virus infection or the derivative of deazapurine
JP2013512859A (en) * 2009-12-03 2013-04-18 大日本住友製薬株式会社 Imidazoquinoline acting through a toll-like receptor (TLR)
US20110150836A1 (en) * 2009-12-22 2011-06-23 Gilead Sciences, Inc. Methods of treating hbv and hcv infection
JP6026405B2 (en) 2010-04-30 2016-11-16 テロルメディクス エセアー Phospholipid drug analogues
WO2011134669A1 (en) * 2010-04-30 2011-11-03 Telormedix Sa Methods for inducing an immune response
US9050319B2 (en) 2010-04-30 2015-06-09 Telormedix, Sa Phospholipid drug analogs
MX2012013713A (en) 2010-05-26 2013-01-28 Selecta Biosciences Inc Nanocarrier compositions with uncoupled adjuvant.
WO2012038058A1 (en) 2010-09-21 2012-03-29 Telormedix Sa Treatment of conditions by toll-like receptor modulators
US9994443B2 (en) 2010-11-05 2018-06-12 Selecta Biosciences, Inc. Modified nicotinic compounds and related methods
JP5978225B2 (en) 2010-12-16 2016-08-24 大日本住友製薬株式会社 Imidazo [4,5-c] quinolin-1-yl derivatives useful for therapy
US8895570B2 (en) 2010-12-17 2014-11-25 Astrazeneca Ab Purine derivatives
US20120231023A1 (en) * 2011-03-08 2012-09-13 Baylor Research Institute Novel Vaccine Adjuvants Based on Targeting Adjuvants to Antibodies Directly to Antigen-Presenting Cells
MX2014000872A (en) * 2011-07-22 2014-07-28 Pawel Kalinski Tumor selective chemokine modulation.
US10112946B2 (en) 2011-07-22 2018-10-30 Glaxosmithkline Llc Composition
AR092198A1 (en) 2012-08-24 2015-04-08 Glaxosmithkline Llc DERIVATIVES OF PIRAZOLOPIRIMIDINAS
WO2014052828A1 (en) 2012-09-27 2014-04-03 The Regents Of The University Of California Compositions and methods for modulating tlr4
AU2013348217B2 (en) 2012-11-20 2016-10-06 Glaxosmithkline Llc Novel compounds
ME02867B (en) 2012-11-20 2018-04-20 Glaxosmithkline Llc Novel compounds
WO2014081643A1 (en) 2012-11-20 2014-05-30 Glaxosmithkline Llc Novel compounds
JP6356160B2 (en) * 2013-02-25 2018-07-11 ザ スクリプス リサーチ インスティテュート Neoceptin: small molecule adjuvant
US11116774B2 (en) 2014-07-11 2021-09-14 Gilead Sciences, Inc. Modulators of toll-like receptors for the treatment of HIV
US9918959B2 (en) 2014-08-06 2018-03-20 The Board Of Regents Of The University Of Texas System TLR-independent small molecule adjuvants
WO2016023511A1 (en) 2014-08-15 2016-02-18 正大天晴药业集团股份有限公司 Pyrrolopyrimidine compounds used as tlr7 agonist
AU2015318061B2 (en) 2014-09-16 2018-05-17 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
EA035116B1 (en) 2015-11-05 2020-04-29 Чиа Тай Тяньцин Фармасьютикал Груп Ко., Лтд. 7-(thiazol-5-yl)pyrrolopyrimidine as tlr7 receptor agonist
CN107043380A (en) 2016-02-05 2017-08-15 正大天晴药业集团股份有限公司 A kind of maleate of TLR7 activators, its crystal formation C, crystal formation D, crystal formation E, preparation method and purposes
CN107043379A (en) 2016-02-05 2017-08-15 正大天晴药业集团股份有限公司 A kind of crystal formation A, its preparation method and the medical usage of TLR7 activators
KR102590454B1 (en) 2016-07-07 2023-10-17 더 보드 어브 트러스티스 어브 더 리랜드 스탠포드 주니어 유니버시티 Antibody-Adjuvant Conjugate
EP3554550A1 (en) 2016-12-13 2019-10-23 Bolt Biotherapeutics, Inc. Antibody adjuvant conjugates
ES2939384T3 (en) 2017-04-14 2023-04-21 Bolt Biotherapeutics Inc Immunoconjugate synthesis method
JP2021035910A (en) * 2017-11-01 2021-03-04 大日本住友製薬株式会社 Substituted purine compound
WO2019196918A1 (en) * 2018-04-13 2019-10-17 罗欣药业(上海)有限公司 Five-membered heterocyclo-pyrimidine compound, pharmaceutical composition and use thereof
US20210154316A1 (en) 2018-05-17 2021-05-27 Bolt Biotherapeutics, Inc. Immunoconjugates
CN109608462B (en) * 2018-12-15 2021-11-23 华南理工大学 7-alkyl-9-alkoxy/thiopurine-8-ketone compound, and synthesis method and application thereof in medicines
CN113993549A (en) 2019-03-15 2022-01-28 博尔特生物治疗药物有限公司 Immunoconjugates targeting HER2
US20220347312A1 (en) 2019-09-04 2022-11-03 Bolt Biotherapeutics, Inc. Immunoconjugate Synthesis Method
CN114787165B (en) * 2020-09-27 2023-02-28 上海维申医药有限公司 Macrocyclic TLR7 agonist, preparation method thereof, pharmaceutical composition and application thereof
CN116600830A (en) 2020-10-21 2023-08-15 卢布尔雅那大学 Conjugated TLR7 and NOD2 agonists

Citations (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0145340A2 (en) 1983-11-18 1985-06-19 Riker Laboratories, Inc. 1H-Imidazo[4,5-c]quinolines and 1H-imidazo[4,5-c]quinolin-4-amines
US4689338A (en) 1983-11-18 1987-08-25 Riker Laboratories, Inc. 1H-Imidazo[4,5-c]quinolin-4-amines and antiviral use
US4929624A (en) 1989-03-23 1990-05-29 Minnesota Mining And Manufacturing Company Olefinic 1H-imidazo(4,5-c)quinolin-4-amines
US4938949A (en) 1988-09-12 1990-07-03 University Of New York Treatment of damaged bone marrow and dosage units therefor
EP0394026A1 (en) 1989-04-20 1990-10-24 Riker Laboratories, Inc. Formulation containing an imidazo[4,5-c]quinolin derivative
US5037986A (en) 1989-03-23 1991-08-06 Minnesota Mining And Manufacturing Company Olefinic 1H-imidazo[4,5-c]quinolin-4-amines
WO1992015581A1 (en) 1991-03-01 1992-09-17 Minnesota Mining And Manufacturing Company PROCESS FOR IMIDAZO[4,5-c]QUINOLIN-4-AMINES
EP0553202A1 (en) 1990-10-05 1993-08-04 Minnesota Mining & Mfg Process for the preparation of imidazo[4,5-c]quinolin-4-amines.
US5238944A (en) 1988-12-15 1993-08-24 Riker Laboratories, Inc. Topical formulations and transdermal delivery systems containing 1-isobutyl-1H-imidazo[4,5-c]quinolin-4-amine
WO1993020847A1 (en) 1992-04-16 1993-10-28 Minnesota Mining And Manufacturing Company Vaccine adjuvant
US5352784A (en) 1993-07-15 1994-10-04 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines
US5395937A (en) 1993-01-29 1995-03-07 Minnesota Mining And Manufacturing Company Process for preparing quinoline amines
US5624677A (en) 1995-06-13 1997-04-29 Pentech Pharmaceuticals, Inc. Controlled release of drugs delivered by sublingual or buccal administration
US5627281A (en) 1993-07-15 1997-05-06 Minnesota Mining And Manufacturing Company Intermediate compounds of fused cycloalkylimidazopyridines
US5693811A (en) 1996-06-21 1997-12-02 Minnesota Mining And Manufacturing Company Process for preparing tetrahdroimidazoquinolinamines
US5736553A (en) 1988-12-15 1998-04-07 Riker Laboratories, Inc. Topical formulations and transdermal delivery systems containing 1-isobutyl-1H-imidazo 4,5-C!quinolin-4-amine
US5741908A (en) 1996-06-21 1998-04-21 Minnesota Mining And Manufacturing Company Process for reparing imidazoquinolinamines
WO1998017279A1 (en) 1996-10-25 1998-04-30 Minnesota Mining And Manufacturing Company Immune response modifier compounds for treatment of th2 mediated and related diseases
WO1998048805A1 (en) 1997-04-25 1998-11-05 Sumitomo Pharmaceuticals Company, Limited Pharmaceutical composition for supressing type 2 helper t cell immune response
WO1999028321A1 (en) 1997-11-28 1999-06-10 Sumitomo Pharmaceuticals Company, Limited Novel heterocyclic compounds
JPH11193282A (en) 1997-12-26 1999-07-21 Sumitomo Pharmaceut Co Ltd Heterocyclic compound
US6038505A (en) 1996-09-12 2000-03-14 Siemens Aktiengesellschaft Method of controlling the drive train of a motor vehicle, and integrated drive train control system
WO2000043394A1 (en) 1999-01-26 2000-07-27 Ústav Experimentální Botaniky Av Cr Substituted nitrogen heterocyclic derivatives and pharmaceutical use thereof
US6245776B1 (en) 1999-01-08 2001-06-12 3M Innovative Properties Company Formulations and methods for treatment of mucosal associated conditions with an immune response modifier
WO2001044260A2 (en) 1999-12-17 2001-06-21 Ariad Pharmaceuticals, Inc. Novel purines
WO2001044259A1 (en) 1999-12-17 2001-06-21 Ariad Pharmaceuticals, Inc. Purine derivatives
WO2001049688A1 (en) 2000-01-07 2001-07-12 Universitaire Instelling Antwerpen Purine derivatives, process for their preparation and use thereof
US6333331B1 (en) 1994-08-01 2001-12-25 The United States Of America As Represented By The Department Of Health And Human Services Substituted O6-benzylguanines
WO2002024225A1 (en) 2000-09-20 2002-03-28 Glaxo Group Limited Use of immidazoquinolinamines as adjuvants in dna vaccination
US6372725B1 (en) 1995-02-16 2002-04-16 Harald Zilch Specific lipid conjugates to nucleoside diphosphates and their use as drugs
US20020127224A1 (en) 2001-03-02 2002-09-12 James Chen Use of photoluminescent nanoparticles for photodynamic therapy
US6486168B1 (en) 1999-01-08 2002-11-26 3M Innovative Properties Company Formulations and methods for treatment of mucosal associated conditions with an immune response modifier
US20020193595A1 (en) 2001-04-09 2002-12-19 Daniel Chu Novel guanidino compounds
WO2003077944A1 (en) 2002-03-19 2003-09-25 Glaxo Group Limited Improvements in vaccination
EP1386923A1 (en) 2001-04-17 2004-02-04 Sumitomo Pharmaceuticals Company, Limited Novel adenine derivatives
US20040023211A1 (en) 2000-09-15 2004-02-05 Kees Groen System and method for optimizing drug theraphy for the treatment of diseases
US6706728B2 (en) 1999-01-08 2004-03-16 3M Innovative Properties Company Systems and methods for treating a mucosal surface
WO2004029054A1 (en) 2002-09-27 2004-04-08 Sumitomo Pharmaceuticals Company, Limited Novel adenine compound and use thereof
US6734187B1 (en) 1997-11-12 2004-05-11 Mitsubishi Chemical Corporation Purine derivatives and medicaments comprising the same as active ingredient
US6733764B2 (en) 2000-06-14 2004-05-11 Alain Martin Immunostimulator anti-cancer compounds and methods for their use in the treatment of cancer
US20040091491A1 (en) * 2002-08-15 2004-05-13 3M Innovative Properties Company Immunostimulatory compositions and methods of stimulating an immune response
JP2004137157A (en) 2002-10-16 2004-05-13 Sumitomo Pharmaceut Co Ltd Medicine comprising new adenine derivative as active ingredient
US20040209899A1 (en) 2001-06-29 2004-10-21 Venkata Palle A2B adenosine receptor antagonists
US20040265351A1 (en) 2003-04-10 2004-12-30 Miller Richard L. Methods and compositions for enhancing immune response
US20050004144A1 (en) 2003-04-14 2005-01-06 Regents Of The University Of California Combined use of IMPDH inhibitors with toll-like receptor agonists
JP2005505504A (en) 2001-05-18 2005-02-24 サーナ・セラピューティクス・インコーポレイテッド Conjugates and compositions for cellular delivery
JP2005046160A (en) 1996-10-25 2005-02-24 Gilead Sciences Inc Vascular endothelial growth factor (vegf) nucleic acid ligand complexes
US20050049263A1 (en) 2001-10-30 2005-03-03 Kasibhatla Srinivas Rao Purine analogs having hsp90-inhibiting activity
US20050054590A1 (en) 2003-09-05 2005-03-10 Averett Devron R. Administration of TLR7 ligands and prodrugs thereof for treatment of infection by hepatitis C virus
US20050059613A1 (en) 2003-07-08 2005-03-17 Bahram Memarzadeh Compositions and methods for the enhanced uptake of therapeutic agents through the bladder epithelium
JP2005089334A (en) 2003-09-12 2005-04-07 Sumitomo Pharmaceut Co Ltd 8-hydroxyadenine compound
WO2005060966A1 (en) 2003-12-19 2005-07-07 Sanofi Pasteur Immunostimulant composition comprising at least one toll-like receptor 7 or toll-like receptor 8 agonist and a toll-like receptor 4 agonist
WO2005092892A1 (en) 2004-03-26 2005-10-06 Dainippon Sumitomo Pharma Co., Ltd. 8-oxoadenine compound
US20050266067A1 (en) 2004-03-02 2005-12-01 Shiladitya Sengupta Nanocell drug delivery system
US7001609B1 (en) 1998-10-02 2006-02-21 Regents Of The University Of Minnesota Mucosal originated drug delivery systems and animal applications
US20060110746A1 (en) 2004-11-19 2006-05-25 Institut Gustave Roussy Treatment of cancer using TLR3 agonists
WO2006062945A2 (en) 2004-12-07 2006-06-15 University Of Miami Cell-derived microparticles as hemostatic agents for control of hemorrhage and treatment of bleeding disorders
WO2006100226A1 (en) 2005-03-21 2006-09-28 Ferrer Internacional, S. A. Method for making 1-substituted 1h-imidazo[4,5-c]quinolin-4-amine compounds and intermediates therefor
WO2007024707A2 (en) 2005-08-22 2007-03-01 The Regents Of The University Of California Tlr agonists
WO2007034817A1 (en) 2005-09-22 2007-03-29 Dainippon Sumitomo Pharma Co., Ltd. Novel adenine compound
WO2007034917A1 (en) 2005-09-22 2007-03-29 Dainippon Sumitomo Pharma Co., Ltd. Novel adenine compound
WO2007038720A2 (en) 2005-09-27 2007-04-05 Coley Pharmaceutical Gmbh Modulation of tlr-mediated immune responses using adaptor oligonucleotides
US20070100146A1 (en) 2005-11-03 2007-05-03 Trevor Dzwiniel Process for the preparation of imidazo[4,5-c]-quinolin-4-amines
US20070161582A1 (en) 2003-08-08 2007-07-12 Dusan Mijikovic Pharmaceutical compositions and methods for metabolic modulation
US20070173483A1 (en) 2002-10-30 2007-07-26 Conforma Therapeutics Corporation Pyrrolopyrimidines and Related Analogs as HSP90-Inhibitors
US20070190071A1 (en) 2004-03-26 2007-08-16 Dainippon Sumitomo Pharma Co., Ltd. 9-Substituted 8-oxoadenine compound
WO2007142755A2 (en) 2006-05-31 2007-12-13 The Regents Of The University Of California Purine analogs
US20070292418A1 (en) 2005-04-26 2007-12-20 Eisai Co., Ltd. Compositions and methods for immunotherapy
US20080008682A1 (en) 2006-07-07 2008-01-10 Chong Lee S Modulators of toll-like receptor 7
US20080214580A1 (en) 2006-10-04 2008-09-04 Pharmacopeia, Inc. 6-substituted 2-(benzimidazolyl)purine and purinone derivatives for immunosuppression
WO2008115319A2 (en) 2007-02-07 2008-09-25 Regents Of The University Of California Conjugates of synthetic tlr agonists and uses therefor
WO2009005687A1 (en) 2007-06-29 2009-01-08 Gilead Sciences, Inc. Purine derivatives and their use as modulators of toll-like receptor 7
US20090069289A1 (en) 2006-10-04 2009-03-12 Pharmacopeia, Inc. 6-substituted 2-(benzimidazolyl)purine and purinone derivatives for immunosuppression
US20090099212A1 (en) 2007-10-16 2009-04-16 Jeff Zablocki A3 adenosine receptor antagonists
US20090202626A1 (en) 2008-02-07 2009-08-13 Carson Dennis A Treatment of bladder diseases with a tlr7 activator
WO2010093436A2 (en) 2009-02-11 2010-08-19 Carson Dennis A Toll-like receptor modulators and treatment of diseases
US20110098248A1 (en) 2009-10-22 2011-04-28 Gilead Sciences, Inc. Modulators of toll-like receptors
WO2011139348A2 (en) 2010-04-30 2011-11-10 The Regents Of The University Of California Uses of phospholipid conjugates of synthetic tlr7 agonists
US20110319442A1 (en) 2009-02-06 2011-12-29 Telormedix Sa Pharmaceutical compositions comprising imidazoquinolin(amines) and derivatives thereof suitable for local administration
US20120003298A1 (en) 2010-04-30 2012-01-05 Alcide Barberis Methods for inducing an immune response
US20120009247A1 (en) 2010-04-30 2012-01-12 Roberto Maj Phospholipid drug analogs
US20120083473A1 (en) 2010-09-21 2012-04-05 Johanna Holldack Treatment of conditions by toll-like receptor modulators
US20120177681A1 (en) 2010-09-01 2012-07-12 Manmohan Singh Formulation of immunopotentiators

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4559157A (en) 1983-04-21 1985-12-17 Creative Products Resource Associates, Ltd. Cosmetic applicator useful for skin moisturizing
LU84979A1 (en) 1983-08-30 1985-04-24 Oreal COSMETIC OR PHARMACEUTICAL COMPOSITION IN AQUEOUS OR ANHYDROUS FORM WHOSE FATTY PHASE CONTAINS OLIGOMER POLYETHER AND NEW OLIGOMER POLYETHERS
US4820508A (en) 1987-06-23 1989-04-11 Neutrogena Corporation Skin protective composition
US4992478A (en) 1988-04-04 1991-02-12 Warner-Lambert Company Antiinflammatory skin moisturizing composition and method of preparing same

Patent Citations (163)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4689338A (en) 1983-11-18 1987-08-25 Riker Laboratories, Inc. 1H-Imidazo[4,5-c]quinolin-4-amines and antiviral use
EP0310950A1 (en) 1983-11-18 1989-04-12 Riker Laboratories, Inc. Quinoline intermediates for the synthesis of 1H-imidazo[4,5-c]quinolines and 1H-imidazo[4,5-c]quinolin-4-amimes
EP0145340A2 (en) 1983-11-18 1985-06-19 Riker Laboratories, Inc. 1H-Imidazo[4,5-c]quinolines and 1H-imidazo[4,5-c]quinolin-4-amines
US4938949A (en) 1988-09-12 1990-07-03 University Of New York Treatment of damaged bone marrow and dosage units therefor
US5736553A (en) 1988-12-15 1998-04-07 Riker Laboratories, Inc. Topical formulations and transdermal delivery systems containing 1-isobutyl-1H-imidazo 4,5-C!quinolin-4-amine
US5238944A (en) 1988-12-15 1993-08-24 Riker Laboratories, Inc. Topical formulations and transdermal delivery systems containing 1-isobutyl-1H-imidazo[4,5-c]quinolin-4-amine
US4929624A (en) 1989-03-23 1990-05-29 Minnesota Mining And Manufacturing Company Olefinic 1H-imidazo(4,5-c)quinolin-4-amines
EP0389302A1 (en) 1989-03-23 1990-09-26 Riker Laboratories, Inc. Olefinic 1H-imidazo [4,5-c]quinolin-4-amines
US5037986A (en) 1989-03-23 1991-08-06 Minnesota Mining And Manufacturing Company Olefinic 1H-imidazo[4,5-c]quinolin-4-amines
EP0394026A1 (en) 1989-04-20 1990-10-24 Riker Laboratories, Inc. Formulation containing an imidazo[4,5-c]quinolin derivative
EP0553202A1 (en) 1990-10-05 1993-08-04 Minnesota Mining & Mfg Process for the preparation of imidazo[4,5-c]quinolin-4-amines.
US5367076A (en) 1990-10-05 1994-11-22 Minnesota Mining And Manufacturing Company Process for imidazo[4,5-C]quinolin-4-amines
US5175296A (en) 1991-03-01 1992-12-29 Minnesota Mining And Manufacturing Company Imidazo[4,5-c]quinolin-4-amines and processes for their preparation
EP0575549A1 (en) 1991-03-01 1993-12-29 Minnesota Mining & Mfg PROCESS FOR IMIDAZO 4,5-c]QUINOLIN-4-AMINES.
WO1992015581A1 (en) 1991-03-01 1992-09-17 Minnesota Mining And Manufacturing Company PROCESS FOR IMIDAZO[4,5-c]QUINOLIN-4-AMINES
WO1993020847A1 (en) 1992-04-16 1993-10-28 Minnesota Mining And Manufacturing Company Vaccine adjuvant
US6083505A (en) 1992-04-16 2000-07-04 3M Innovative Properties Company 1H-imidazo[4,5-C]quinolin-4-amines as vaccine adjuvants
EP0636031A1 (en) 1992-04-16 1995-02-01 Minnesota Mining & Mfg Vaccine adjuvant.
US5395937A (en) 1993-01-29 1995-03-07 Minnesota Mining And Manufacturing Company Process for preparing quinoline amines
EP0681570A1 (en) 1993-01-29 1995-11-15 Minnesota Mining And Manufacturing Company Process for preparing quinoline amines
EP0708773A1 (en) 1993-07-15 1996-05-01 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines
US5627281A (en) 1993-07-15 1997-05-06 Minnesota Mining And Manufacturing Company Intermediate compounds of fused cycloalkylimidazopyridines
US5352784A (en) 1993-07-15 1994-10-04 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines
US5444065A (en) 1993-07-15 1995-08-22 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines as inducer of interferon α biosynthesis
US5648516A (en) 1994-07-20 1997-07-15 Minnesota Mining And Manufacturing Company Fused cycloalkylimidazopyridines
US6333331B1 (en) 1994-08-01 2001-12-25 The United States Of America As Represented By The Department Of Health And Human Services Substituted O6-benzylguanines
US6372725B1 (en) 1995-02-16 2002-04-16 Harald Zilch Specific lipid conjugates to nucleoside diphosphates and their use as drugs
US5624677A (en) 1995-06-13 1997-04-29 Pentech Pharmaceuticals, Inc. Controlled release of drugs delivered by sublingual or buccal administration
EP0912564A1 (en) 1996-06-21 1999-05-06 Minnesota Mining And Manufacturing Company Process for preparing tetrahydroimidazoquinolinamines
US6624305B2 (en) 1996-06-21 2003-09-23 3M Innovative Properties Company Process for preparing imidazoquinolinamines
EP0912565A1 (en) 1996-06-21 1999-05-06 Minnesota Mining And Manufacturing Company Process for preparing imidazoquinolinamines
US6613902B2 (en) 1996-06-21 2003-09-02 3M Innovative Properties Company Process for preparing imidazoquinolinamines
US6534654B2 (en) 1996-06-21 2003-03-18 3M Innovative Properties Company Process for preparing imidazoquinolinamines
US6437131B1 (en) 1996-06-21 2002-08-20 3M Innovative Properties Company Process for preparing imidazoquinolinamines
US5998619A (en) 1996-06-21 1999-12-07 3M Innovative Properties Company Process for preparing imidazoquinolinamines
US5741908A (en) 1996-06-21 1998-04-21 Minnesota Mining And Manufacturing Company Process for reparing imidazoquinolinamines
US6150523A (en) 1996-06-21 2000-11-21 3M Innovative Properties Company Process for preparing imidazoquinolinamines
US5693811A (en) 1996-06-21 1997-12-02 Minnesota Mining And Manufacturing Company Process for preparing tetrahdroimidazoquinolinamines
US6897314B2 (en) 1996-06-21 2005-05-24 3M Innovative Properties Company Process for preparing imidazoquinolinamines
US6038505A (en) 1996-09-12 2000-03-14 Siemens Aktiengesellschaft Method of controlling the drive train of a motor vehicle, and integrated drive train control system
US6200592B1 (en) 1996-10-25 2001-03-13 3M Innovative Properties Company Immine response modifier compounds for treatment of TH2 mediated and related diseases
JP2005046160A (en) 1996-10-25 2005-02-24 Gilead Sciences Inc Vascular endothelial growth factor (vegf) nucleic acid ligand complexes
US6696076B2 (en) 1996-10-25 2004-02-24 3M Innovative Properties Company Immune response modifier compounds for treatment of TH2 mediated and related diseases
US6610319B2 (en) 1996-10-25 2003-08-26 3M Innovative Properties Company Immune response modifier compounds for treatment of TH2 mediated and related diseases
WO1998017279A1 (en) 1996-10-25 1998-04-30 Minnesota Mining And Manufacturing Company Immune response modifier compounds for treatment of th2 mediated and related diseases
EP0938315A1 (en) 1996-10-25 1999-09-01 Minnesota Mining And Manufacturing Company Immune response modifier compounds for treatment of th2 mediated and related diseases
US6039969A (en) 1996-10-25 2000-03-21 3M Innovative Properties Company Immune response modifier compounds for treatment of TH2 mediated and related diseases
WO1998048805A1 (en) 1997-04-25 1998-11-05 Sumitomo Pharmaceuticals Company, Limited Pharmaceutical composition for supressing type 2 helper t cell immune response
US6734187B1 (en) 1997-11-12 2004-05-11 Mitsubishi Chemical Corporation Purine derivatives and medicaments comprising the same as active ingredient
US6329381B1 (en) 1997-11-28 2001-12-11 Sumitomo Pharmaceuticals Company, Limited Heterocyclic compounds
EP1035123A1 (en) * 1997-11-28 2000-09-13 Sumitomo Pharmaceuticals Company, Limited Novel heterocyclic compounds
WO1999028321A1 (en) 1997-11-28 1999-06-10 Sumitomo Pharmaceuticals Company, Limited Novel heterocyclic compounds
JPH11193282A (en) 1997-12-26 1999-07-21 Sumitomo Pharmaceut Co Ltd Heterocyclic compound
US7001609B1 (en) 1998-10-02 2006-02-21 Regents Of The University Of Minnesota Mucosal originated drug delivery systems and animal applications
US6486168B1 (en) 1999-01-08 2002-11-26 3M Innovative Properties Company Formulations and methods for treatment of mucosal associated conditions with an immune response modifier
US6706728B2 (en) 1999-01-08 2004-03-16 3M Innovative Properties Company Systems and methods for treating a mucosal surface
US6245776B1 (en) 1999-01-08 2001-06-12 3M Innovative Properties Company Formulations and methods for treatment of mucosal associated conditions with an immune response modifier
WO2000043394A1 (en) 1999-01-26 2000-07-27 Ústav Experimentální Botaniky Av Cr Substituted nitrogen heterocyclic derivatives and pharmaceutical use thereof
US6552192B1 (en) 1999-01-26 2003-04-22 Ustau Experimentalni Botaniky Av-Cr Substituted nitrogen heterocyclic derivatives and pharmaceutical use thereof
US20030191086A1 (en) 1999-01-26 2003-10-09 Ustav Experimentalni Botaniky Av Cr Substituted nitrogen heterocyclic derivatives and pharmaceutical use thereof
WO2001044260A2 (en) 1999-12-17 2001-06-21 Ariad Pharmaceuticals, Inc. Novel purines
WO2001044259A1 (en) 1999-12-17 2001-06-21 Ariad Pharmaceuticals, Inc. Purine derivatives
US20030187261A1 (en) 2000-01-07 2003-10-02 Libor Havlicek Purine derivatives, process for their preparation and use thereof
WO2001049688A1 (en) 2000-01-07 2001-07-12 Universitaire Instelling Antwerpen Purine derivatives, process for their preparation and use thereof
US6733764B2 (en) 2000-06-14 2004-05-11 Alain Martin Immunostimulator anti-cancer compounds and methods for their use in the treatment of cancer
US20040023211A1 (en) 2000-09-15 2004-02-05 Kees Groen System and method for optimizing drug theraphy for the treatment of diseases
WO2002024225A1 (en) 2000-09-20 2002-03-28 Glaxo Group Limited Use of immidazoquinolinamines as adjuvants in dna vaccination
US20020127224A1 (en) 2001-03-02 2002-09-12 James Chen Use of photoluminescent nanoparticles for photodynamic therapy
US20050038027A1 (en) 2001-04-09 2005-02-17 Chiron Corporation Novel guanidino compounds
US6716840B2 (en) 2001-04-09 2004-04-06 Chiron Corporation Guanidino compounds
US7189727B2 (en) 2001-04-09 2007-03-13 Chiron Corporation Guanidino compounds
US6960582B2 (en) 2001-04-09 2005-11-01 Chiron Corporation Guanidino compounds
US20020193595A1 (en) 2001-04-09 2002-12-19 Daniel Chu Novel guanidino compounds
US20040248895A1 (en) 2001-04-09 2004-12-09 Daniel Chu Novel guanidino compounds
US20040132748A1 (en) 2001-04-17 2004-07-08 Yoshiaki Isobe Novel adenne derivatives
EP1386923A1 (en) 2001-04-17 2004-02-04 Sumitomo Pharmaceuticals Company, Limited Novel adenine derivatives
US7521454B2 (en) 2001-04-17 2009-04-21 Dainippon Sumitomo Pharma Co., Ltd. Adenine derivatives
US7157465B2 (en) 2001-04-17 2007-01-02 Dainippon Simitomo Pharma Co., Ltd. Adenine derivatives
US20070037832A1 (en) 2001-04-17 2007-02-15 Dainippon Sumitomo Pharma Co., Ltd. Novel adenine derivatives
JP2005505504A (en) 2001-05-18 2005-02-24 サーナ・セラピューティクス・インコーポレイテッド Conjugates and compositions for cellular delivery
US20040209899A1 (en) 2001-06-29 2004-10-21 Venkata Palle A2B adenosine receptor antagonists
US7238700B2 (en) 2001-06-29 2007-07-03 Cv Therapeutics, Inc. A2B adenosine receptor antagonists
US20050049263A1 (en) 2001-10-30 2005-03-03 Kasibhatla Srinivas Rao Purine analogs having hsp90-inhibiting activity
US7241890B2 (en) 2001-10-30 2007-07-10 Conforma Therapeutics Corporation Purine analogs having HSP90-inhibiting activity
US20080125446A1 (en) 2001-10-30 2008-05-29 Conforma Therapeutics Corporation Purine analogs having HSP90-inhibiting activity
WO2003077944A1 (en) 2002-03-19 2003-09-25 Glaxo Group Limited Improvements in vaccination
US20040091491A1 (en) * 2002-08-15 2004-05-13 3M Innovative Properties Company Immunostimulatory compositions and methods of stimulating an immune response
US20060052403A1 (en) 2002-09-27 2006-03-09 Yoshiaki Isobe Novel adenine compound and use thereof
WO2004029054A1 (en) 2002-09-27 2004-04-08 Sumitomo Pharmaceuticals Company, Limited Novel adenine compound and use thereof
EP1550662A1 (en) * 2002-09-27 2005-07-06 Sumitomo Pharmaceuticals Company, Limited Novel adenine compound and use thereof
US7754728B2 (en) 2002-09-27 2010-07-13 Dainippon Sumitomo Pharma Co., Ltd. Adenine compound and use thereof
JP2004137157A (en) 2002-10-16 2004-05-13 Sumitomo Pharmaceut Co Ltd Medicine comprising new adenine derivative as active ingredient
US20070173483A1 (en) 2002-10-30 2007-07-26 Conforma Therapeutics Corporation Pyrrolopyrimidines and Related Analogs as HSP90-Inhibitors
US20040265351A1 (en) 2003-04-10 2004-12-30 Miller Richard L. Methods and compositions for enhancing immune response
US20050004144A1 (en) 2003-04-14 2005-01-06 Regents Of The University Of California Combined use of IMPDH inhibitors with toll-like receptor agonists
US20050059613A1 (en) 2003-07-08 2005-03-17 Bahram Memarzadeh Compositions and methods for the enhanced uptake of therapeutic agents through the bladder epithelium
US20070161582A1 (en) 2003-08-08 2007-07-12 Dusan Mijikovic Pharmaceutical compositions and methods for metabolic modulation
US20050054590A1 (en) 2003-09-05 2005-03-10 Averett Devron R. Administration of TLR7 ligands and prodrugs thereof for treatment of infection by hepatitis C virus
JP2007504232A (en) 2003-09-05 2007-03-01 アナディス ファーマシューティカルズ インク Administration of TLR7 ligand and prodrug thereof for the treatment of hepatitis C virus infection
US7576068B2 (en) 2003-09-05 2009-08-18 Anadys Pharmaceuticals, Inc. Administration of TLR7 ligands and prodrugs thereof for treatment of infection by hepatitis C virus
WO2005025583A2 (en) 2003-09-05 2005-03-24 Anadys Pharmaceuticals, Inc. Tlr7 ligands for the treatment of hepatitis c
US8211863B2 (en) 2003-09-05 2012-07-03 Anadys Pharmaceuticals, Inc. Administration of TLR7 ligands and prodrugs thereof for treatment of infection by hepatitis C virus
JP2005089334A (en) 2003-09-12 2005-04-07 Sumitomo Pharmaceut Co Ltd 8-hydroxyadenine compound
US20070087009A1 (en) 2003-12-19 2007-04-19 Sanofi Pasteur Immunostimulant composition comprising at least one toll-like receptor 7 or toll-like receptor 8 agonist and a toll-like receptor 4 agonist
WO2005060966A1 (en) 2003-12-19 2005-07-07 Sanofi Pasteur Immunostimulant composition comprising at least one toll-like receptor 7 or toll-like receptor 8 agonist and a toll-like receptor 4 agonist
US20050266067A1 (en) 2004-03-02 2005-12-01 Shiladitya Sengupta Nanocell drug delivery system
US20070190071A1 (en) 2004-03-26 2007-08-16 Dainippon Sumitomo Pharma Co., Ltd. 9-Substituted 8-oxoadenine compound
WO2005092892A1 (en) 2004-03-26 2005-10-06 Dainippon Sumitomo Pharma Co., Ltd. 8-oxoadenine compound
US20060110746A1 (en) 2004-11-19 2006-05-25 Institut Gustave Roussy Treatment of cancer using TLR3 agonists
WO2006062945A2 (en) 2004-12-07 2006-06-15 University Of Miami Cell-derived microparticles as hemostatic agents for control of hemorrhage and treatment of bleeding disorders
WO2006100226A1 (en) 2005-03-21 2006-09-28 Ferrer Internacional, S. A. Method for making 1-substituted 1h-imidazo[4,5-c]quinolin-4-amine compounds and intermediates therefor
US20070292418A1 (en) 2005-04-26 2007-12-20 Eisai Co., Ltd. Compositions and methods for immunotherapy
JP2009504803A (en) 2005-08-22 2009-02-05 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア TLR agonist
WO2007024707A3 (en) 2005-08-22 2007-09-20 Univ California Tlr agonists
US20090324551A1 (en) 2005-08-22 2009-12-31 The Regents Of The University Of California Office Of Technology Transfer Tlr agonists
WO2007024707A2 (en) 2005-08-22 2007-03-01 The Regents Of The University Of California Tlr agonists
WO2007034917A1 (en) 2005-09-22 2007-03-29 Dainippon Sumitomo Pharma Co., Ltd. Novel adenine compound
US20090118263A1 (en) 2005-09-22 2009-05-07 Dainippon Sumitomo Pharma Co., Ltd. Novel Adenine Compound
US20090105212A1 (en) 2005-09-22 2009-04-23 Dainippon Sumitomo Pharma Co., Ltd. a corporation of Japan Novel adenine compound
WO2007034817A1 (en) 2005-09-22 2007-03-29 Dainippon Sumitomo Pharma Co., Ltd. Novel adenine compound
EP1939202A1 (en) 2005-09-22 2008-07-02 Dainippon Sumitomo Pharma Co., Ltd. Novel adenine compound
JP2009510096A (en) 2005-09-27 2009-03-12 コーリー ファーマシューティカル ゲーエムベーハー Modulation of TLR-mediated immune responses using adapter oligonucleotides
WO2007038720A2 (en) 2005-09-27 2007-04-05 Coley Pharmaceutical Gmbh Modulation of tlr-mediated immune responses using adaptor oligonucleotides
US20070100146A1 (en) 2005-11-03 2007-05-03 Trevor Dzwiniel Process for the preparation of imidazo[4,5-c]-quinolin-4-amines
US20130190494A1 (en) 2006-05-31 2013-07-25 The Regents Of The University Of California Purine analogs
WO2007142755A3 (en) 2006-05-31 2008-10-02 Univ California Purine analogs
WO2007142755A2 (en) 2006-05-31 2007-12-13 The Regents Of The University Of California Purine analogs
AU2007257423B2 (en) 2006-05-31 2012-02-09 The Regents Of The University Of California Purine analogs
US8846697B2 (en) 2006-05-31 2014-09-30 The Regents Of The University Of California Purine analogs
US20110098294A1 (en) 2006-05-31 2011-04-28 Carson Dennis A Purine analogs
US20090202484A1 (en) 2006-07-07 2009-08-13 Gilead Sciences, Inc. Modulators of toll-like receptor 7
US20080008682A1 (en) 2006-07-07 2008-01-10 Chong Lee S Modulators of toll-like receptor 7
US20080214580A1 (en) 2006-10-04 2008-09-04 Pharmacopeia, Inc. 6-substituted 2-(benzimidazolyl)purine and purinone derivatives for immunosuppression
US20090069289A1 (en) 2006-10-04 2009-03-12 Pharmacopeia, Inc. 6-substituted 2-(benzimidazolyl)purine and purinone derivatives for immunosuppression
WO2008115319A3 (en) 2007-02-07 2009-03-05 Dennis A Carson Conjugates of synthetic tlr agonists and uses therefor
US20140302120A1 (en) 2007-02-07 2014-10-09 The Regents Of The University Of California Conjugates of synthetic tlr agonists and uses therefor
US8790655B2 (en) 2007-02-07 2014-07-29 The Regents Of The University Of California Conjugates of synthetic TLR agonists and uses therefor
WO2008115319A2 (en) 2007-02-07 2008-09-25 Regents Of The University Of California Conjugates of synthetic tlr agonists and uses therefor
US8357374B2 (en) 2007-02-07 2013-01-22 The Regents Of The University Of California Conjugates of synthetic TLR agonists and uses therefor
US20120148660A1 (en) 2007-02-07 2012-06-14 Regents Of The University Of California, San Diego Ucsd Technology Transfer Office Conjugates of synthetic tlr agonists and uses therefor
HK1138767A1 (en) 2007-02-07 2010-09-03 Univ California Conjugates of synthetic tlr agonists and uses therefor
US20130156807A1 (en) 2007-02-07 2013-06-20 The Regents Of The University Of California Conjugates of synthetic tlr agonists and uses therefor
WO2009005687A1 (en) 2007-06-29 2009-01-08 Gilead Sciences, Inc. Purine derivatives and their use as modulators of toll-like receptor 7
US7968544B2 (en) 2007-06-29 2011-06-28 Gilead Sciences, Inc. Modulators of toll-like receptor 7
US20090047249A1 (en) 2007-06-29 2009-02-19 Micheal Graupe Modulators of toll-like receptor 7
US20090099212A1 (en) 2007-10-16 2009-04-16 Jeff Zablocki A3 adenosine receptor antagonists
WO2009099650A3 (en) 2008-02-07 2009-10-22 Carson Dennis A Treatment of bladder diseases with a tlr7 activator
US20090202626A1 (en) 2008-02-07 2009-08-13 Carson Dennis A Treatment of bladder diseases with a tlr7 activator
WO2009099650A2 (en) 2008-02-07 2009-08-13 Carson Dennis A Treatment of bladder diseases with a tlr7 activator
WO2009099650A4 (en) 2008-02-07 2010-01-14 Carson Dennis A Treatment of bladder diseases with a tlr7 activator
US20110319442A1 (en) 2009-02-06 2011-12-29 Telormedix Sa Pharmaceutical compositions comprising imidazoquinolin(amines) and derivatives thereof suitable for local administration
EP2396328A2 (en) 2009-02-11 2011-12-21 The Regents of The University of California Toll-like receptor modulators and treatment of diseases
US8729088B2 (en) 2009-02-11 2014-05-20 The Regents Of The University Of California Toll-like receptor modulators and treatment of diseases
US20100210598A1 (en) 2009-02-11 2010-08-19 Regents Of The University Of California, San Diego Toll-like receptor modulators and treatment of diseases
JP2012517428A (en) 2009-02-11 2012-08-02 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Toll-like receptor modulators and disease treatment
WO2010093436A2 (en) 2009-02-11 2010-08-19 Carson Dennis A Toll-like receptor modulators and treatment of diseases
US20110098248A1 (en) 2009-10-22 2011-04-28 Gilead Sciences, Inc. Modulators of toll-like receptors
JP2013525431A (en) 2010-04-30 2013-06-20 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Use of phospholipid conjugates of synthetic TLR7 agonists
US20120009247A1 (en) 2010-04-30 2012-01-12 Roberto Maj Phospholipid drug analogs
US20120003298A1 (en) 2010-04-30 2012-01-05 Alcide Barberis Methods for inducing an immune response
WO2011139348A2 (en) 2010-04-30 2011-11-10 The Regents Of The University Of California Uses of phospholipid conjugates of synthetic tlr7 agonists
US20120177681A1 (en) 2010-09-01 2012-07-12 Manmohan Singh Formulation of immunopotentiators
US20120083473A1 (en) 2010-09-21 2012-04-05 Johanna Holldack Treatment of conditions by toll-like receptor modulators

Non-Patent Citations (250)

* Cited by examiner, † Cited by third party
Title
"8H-Purin-8-one, 6-amino-2-(butylthio)-7,9-dihydro-9-(phenylmethyl)-", CAS Registry No. 226906-70-3.
"8H-Purin-8-one, 6-amino-2-(cyclohexylthio)-7,9-dihydro-9-(phenylmethyl)-", CAS Registry No. 226906-76-9.
"8H-Purin-8-one, 6-amino-2-(ethylthio)-7,9-dihydro-9-(phenylmethyl)-", CAS Registry No. 226906-67-8.
"8H-Purin-8-one, 6-amino-7,9-dihydro-2-(methylthio)-9-(phenylmethyl)-", CAS Registry No. 226906-66-7.
"8H-Purin-8-one, 6-amino-7,9-dihydro-2-(pentylthio)-9-(phenylmethyl)-", CAS Registry No. 226906-73-6.
"8H-Purin-8-one, 6-amino-7,9-dihydro-2-[(1-methylethyl)thio]-9-(phenylmethyl)-", CAS Registry No. 226906-69-0.
"8H-Purin-8-one, 6-amino-7,9-dihydro-2-[(1-methylpropyl)thio]-9-(phenylmethyl)-", CAS Registry No. 226906-72-5.
"8H-Purin-8-one, 6-amino-7,9-dihydro-2-[(2-methylbutyl)thio]-9-(phenylmethyl)-", CAS Registry No. 226906-75-8.
"8H-Purin-8-one, 6-amino-7,9-dihydro-2-[(2-methylpropyl)thio]-9-(phenylmethyl)-", CAS Registry No. 226906-71-4.
"8H-Purin-8-one, 6-amino-7,9-dihydro-2-[(3-methylbutyl)thio]-9-(phenylmethyl)-", CAS Registry No. 226906-74-7.
"8H-Purin-8-one, 6-amino-7,9-dihydro-9-(phenylmethyl)-2-(phenylthio)-", CAS Registry No. 226906-77-0.
"8H-Purin-8-one, 6-amino-7,9-dihydro-9-(phenylmethyl)-2-(propylthio)-", CAS Registry No. 226906-68-9.
"Aromatic Ions (Chemgapedia)", [online]. [retrieved on Dec. 3, 2012]. Retrieved From Internet: <URL: http://www.chemgapedia.de/vsengine/vlu/vsc/en/ch/12/oc/vlu-organik/aromaten/aromaten/aromaten-gesamt.vlu/Page/vsc/en/ch/12/oc/aromaten/aromaten/ar-ionen/ar-ionen.vscml.html>, (2012), 2 pgs.
"Australia Application Serial No. 2008227128, First Examiner Report mailed Jul. 6, 2012", 2 pgs.
"Australian Application Serial No. 2006283524, Office Action mailed Aug. 3, 2011", 4 pgs.
"Australian Application Serial No. 2006283524, Office Action mailed Mar. 27, 2008", 1 pg.
"Australian Application Serial No. 2006283524, Preliminary Amendment mailed Mar. 3, 2008", 18 pgs.
"Australian Application Serial No. 2006283524, Response filed Aug. 2, 2012 to Examiner Report mailed Aug. 3, 2011", 34 pgs.
"Australian Application Serial No. 2006283524, Response filed May 19, 2008 to Office Action mailed Mar. 27, 2008", 10 pgs.
"Australian Application Serial No. 2007257423, Examiner Report mailed Jun. 6, 2011", 2 pgs.
"Australian Application Serial No. 2007257423, First Examiner Report mailed Sep. 22, 2010", 4 Pgs.
"Australian Application Serial No. 2007257423, Office Action mailed Oct. 20, 2011", 2 pgs.
"Australian Application Serial No. 2007257423, Response filed Dec. 19, 2011 to Office Action mailed Oct. 20, 2011", 5 pgs.
"Australian Application Serial No. 2007257423, Response filed May 31, 2011 to First Examiner Report mailed Sep. 22, 2010", 16 pgs.
"Australian Application Serial No. 2007257423, Response filed Sep. 13, 2011 to Examination Report mailed Jun. 6, 2011", 12 pgs.
"Australian Application Serial No. 2008227128, Preliminary Amendment filed Sep. 7, 2009", 45 pgs.
"Brazilian Application Serial No. PI 0807196-9, Amendment filed Mar. 2, 2011", 13 pgs.
"Canadian Application Serial No. 2,620,182, Office Action mailed Aug. 24, 2012", 5 pgs.
"Canadian Application Serial No. 2,653,941, Office Action mailed Aug. 23, 2010", 5 pgs.
"Canadian Application Serial No. 2,653,941, Office Action mailed Feb. 8, 2012", 2 pgs.
"Canadian Application Serial No. 2,653,941, Office Action May 10, 2011", 3 pgs.
"Canadian Application Serial No. 2,653,941, Response filed Aug. 2, 2012 to Office Action mailed Feb. 8, 2012", 7 pgs.
"Canadian Application Serial No. 2,653,941, Response filed Feb. 23, 2011 to Office Action mailed Aug. 23, 2010", 20 pgs.
"Canadian Application Serial No. 2,653,941, Response filed Nov. 9, 2011 to Office Action mailed May 10, 2011", 15 pgs.
"Canadian Application Serial No. 2,677,733, Voluntary Amendment filed Aug. 7, 2009", 45 pgs.
"Chinese Application Serial No. 200680038761.X, Office Action mailed Apr. 14, 2010", with English translation, 9 pgs.
"Chinese Application Serial No. 200680038761.X, Office Action mailed Jun. 23, 2011", (w/ English Translation), 9 pgs.
"Chinese Application Serial No. 200680038761.X, Office Action mailed Mar. 22, 2012", 9 pgs.
"Chinese Application Serial No. 200680038761.X, Office Action Response Filed Oct. 29, 2010", with English translation of amended claims, 22 pgs.
"Chinese Application Serial No. 200680038761.X, Response filed Jul. 6, 2012 to Action mailed Mar. 22, 2012", 9 pgs.
"Chinese Application Serial No. 200680038761.X, Response filed Sep. 7, 2011 to Office Action mailed Jun. 23, 2011", (w/ English Translation of Amended Claims), 19 pgs.
"Chinese Application Serial No. 200880011525.8, Office Action mailed Jan. 30, 2012", English Translation Only, 6 pgs.
"Chinese Application Serial No. 200880011525.8, Office Action mailed Jul. 5, 2012", 14 pgs.
"Chinese Application Serial No. 200880011525.8, Office Action mailed Oct. 16, 2012", 13 pgs.
"Chinese Application Serial No. 200880011525.8, Response filed Feb. 27, 2013 to Office Action mailed Oct. 16, 2012", (w/ English Translation of Amended Claims), 10 pgs.
"Chinese Application Serial No. 200880011525.8, Response filed Jun. 13, 2012 to Office Action mailed Jan. 30, 2012", 14 pgs.
"Chinese Application Serial No. 200880011525.8, Response filed Sep. 20, 2012 to Office action Mailed Jul. 5, 2012", 12 pgs.
"Chinese Application Serial No. 200880011525.8, Voluntary Amendment filed Dec. 2, 2010", (w/ English Translation of Claims), 12 pgs.
"Chinese Application Serial No. 200980112411.7, Office Action Mailed Feb. 2, 2012", w/ English Translation, 9 pgs.
"Chinese Application Serial No. 200980112411.7, Response filed Aug. 15, 2012 to Office Action mailed Feb. 2, 2012", (w/ English Translation of Amended Claims), 70 pgs.
"Chinese Application Serial No. 200980112411.7, Voluntary Amendment filed Jan. 31, 2011", (w/ English Translation of Claims), 74 pgs.
"Definition: Micelle", Merriam-Webster, [Online]. Retrieved from the Internet: <URL:http://www.merriam-webster.com/dictionary/micelle>, (Accessed on Jun. 25, 2014), 1 pg.
"Eurasian Application Serial No. 200901078, Office Action mailed Apr. 2, 2012", w/English Translation, 3 pgs.
"Eurasian Application Serial No. 200901078, Office Action mailed Jan. 29, 2013", (w/ English Translation), 4 pgs.
"Eurasian Application Serial No. 200901078, Office Action mailed May 26, 2011", 5 pgs.
"Eurasian Application Serial No. 200901078, Office Action mailed Sep. 18, 2012", 4 pgs.
"Eurasian Application Serial No. 200901078, Office Action mailed Sep. 21, 2011", (w/ English Translation), 4 pgs.
"Eurasian Application Serial No. 200901078, Response filed Jan. 16, 2013 to Office Action mailed Sep. 18, 2012", (w/ English Translation of Claims), 68 pgs.
"Eurasian Application Serial No. 200901078, Response filed Jul. 29, 2013 to Office Action mailed Jan. 29, 2013", (w/ English Translation of Claims), 138 pgs.
"Eurasian Application Serial No. 200901078, Response filed Sep. 13, 2011", 13 pgs.
"Eurasian Application Serial No. 201001264, Office Action mailed Sep. 12, 2012", 1 pg.
"Eurasian Patent Application Serial No. 200901078, Response filed Aug. 2, 2012 to Office Action mailed Apr. 2, 2012", 2 pgs.
"Eurasian Patent Application Serial No. 200901078, Response filed Mar. 21, 2012 to Office Action mailed Sep. 21, 2011", 8 pgs.
"European Application Serial No. 06813535.9, Examination Notification Art. 94(3) mailed Sep. 24, 2013", 4 pgs.
"European Application Serial No. 06813535.9, Extended Search Report mailed Oct. 24, 2011", 6 pgs.
"European Application Serial No. 06813535.9, Response filed May 14, 2012 to Extended Search Report mailed Oct. 24, 2011", 18 pgs.
"European Application Serial No. 06813535.9, Voluntary Amendment filed Apr. 22, 2008", 9 pgs.
"European Application Serial No. 07755916.9, Examination Notification Art. 94(3) mailed Aug. 15, 2013", 4 pgs.
"European Application Serial No. 07755916.9, Office Action mailed Nov. 11, 2011", 1 pg.
"European Application Serial No. 07755916.9, Response filed May 18, 2012 to Extended Search Report mailed Oct. 25, 2011", 11 pgs.
"European Application Serial No. 07755916.9, Supplemental Search Report mailed Oct. 25, 2011", 9 pgs.
"European Application Serial No. 08799591.6, Examination Notification Art. 94(3) mailed May 17, 2011", 5 pgs.
"European Application Serial No. 08799591.6, Office Action mailed Jun. 4, 2010", 4 pgs.
"European Application Serial No. 08799591.6, Office Action mailed May 21, 2012", 4 pgs.
"European Application Serial No. 08799591.6, Office Action Response Dated Sep. 20, 2012", 31 Pgs.
"European Application Serial No. 08799591.6, Response filed Dec. 2, 2010 to Office Action mailed Jun. 4, 2010", 20 pgs.
"European Application Serial No. 08799591.6, Response filed Nov. 22, 2011 to Office Action mailed May 17, 2011", 26 pgs.
"European Application Serial No. 09709019.5, Extended European Search Report mailed Feb. 15, 2011", 8 pgs.
"European Application Serial No. 12004181.9, Communication mailed Oct. 22, 2012", 2 pgs.
"European Application Serial No. 12004181.9, Communication pursuant to Rule 112(1) EPC mailed May 31, 2013", 1 pg.
"European Application Serial No. 12004181.9, Examination Notification Art. 94(3) mailed Sep. 2, 2013", 5 pgs.
"European Application Serial No. 12004181.9, Extended EP Search Report mailed Sep. 13, 2012", 8 pgs.
"European Application Serial No. 12004181.9, Response filed Jul. 31, 2013 to Communication pursuant to Rule 112(1) EPC mailed May 31, 2013 and Communication mailed Oct. 22, 2012", 9 pgs.
"I. Pharmaceutical Importance of Crystallin Hydrates", [online]. [retrieved on May 30, 2008]. Retrieved from the Internet: , (2008), 126-127.
"I. Pharmaceutical Importance of Crystallin Hydrates", [online]. [retrieved on May 30, 2008]. Retrieved from the Internet: <URL: http://www.netlibrary.com/nlreader.dll?bookid=12783&filename=Page-126. html>, (2008), 126-127.
"Indian Application Serial No. 2064/delnp/2008, Examination Report mailed Aug. 21, 2012", 5 pgs.
"Indian Application Serial No. 5675/DELNP/2009, Voluntary Amendment filed Feb. 18, 2011", 7 pgs.
"International Application Serial No. PCT/US06/32371, International Preliminary Report on Patentability mailed Mar. 6, 2008", 6 pgs.
"International Application Serial No. PCT/US06/32371, International Search Report mailed Jul. 23, 2007", 3 pgs.
"International Application Serial No. PCT/US06/32371, Written Opinion mailed Jul. 23, 2007", 6 pgs.
"International Application Serial No. PCT/US2007/009840, International Preliminary Report on Patentability mailed Dec. 18, 2008", 9 pgs.
"International Application Serial No. PCT/US2008/001631, International Preliminary Examination Report mailed Aug. 20, 2009", 12 pgs.
"International Application Serial No. PCT/US2008/001631, International Search Report mailed Jan. 21, 2009", 6 pgs.
"International Application Serial No. PCT/US2008/001631, Written Opinion mailed Jan. 21, 2009", 9 pgs.
"International Application Serial No. PCT/US2009/000771, International Search Report mailed Aug. 28, 2009", 4 pgs.
"International Application Serial No. PCT/US2009/000771, Written Opinion mailed Aug. 28, 2009", 5 pgs.
"International Application Serial No. PCT/US2010/000369, International Preliminary Report on Patentability dated Jun. 28, 2011", 13 pgs.
"International Application Serial No. PCT/US2010/000369, International Search Report mailed Sep. 21, 2010", 6 pgs.
"International Application Serial No. PCT/US2010/000369, Partial International Search Report mailed Jul. 5, 2010", 6 pgs.
"International Application Serial No. PCT/US2010/000369, Written Opinion mailed Feb. 11, 2010", 9 pgs.
"International Application Serial No. PCT/US2010/000369, Written Opinion mailed Sep. 21, 2010", 9 pgs.
"Israeli Application Serial No. 200240, Examiner Report mailed Aug. 28, 2012", 4 pgs.
"Israeli Application Serial No. 200240, Examiner Report mailed Aug. 5, 2013", (English Translation), 3 pgs.
"Israeli Application Serial No. 200240, Response filed Dec. 17, 2012 to Examiner Report mailed Aug. 28, 2012", (w/ English Translation of Claims), 4 pgs.
"Israeli Application Serial No. 200240, Response filed Nov. 25, 2013 to Examiner Report mailed Aug. 5, 2013", 8 pgs.
"Japanese Application No. 2010-545884, Voluntary Amendment filed Oct. 7, 2010", 65 pgs.
"Japanese Application Serial No. 2008-528017, Office Action mailed May 22, 2012", 7 pgs.
"Japanese Application Serial No. 2008-528017, Preliminary Amendment filed Aug. 12, 2009", 26 pgs.
"Japanese Application Serial No. 2008-528017, Response filed Nov. 20, 2012 to Office Action mailed May 22, 2012", (w/ English Translation of Amended Claims), 19 pgs.
"Japanese Application Serial No. 2009-549102, Office Action mailed May 29, 2013", (w/ English Translation), 5 pgs.
"Japanese Application Serial No. 2009-549102, Office Action mailed Oct. 12, 2012", 3 pgs.
"Japanese Application Serial No. 2009-549102, Response filed Aug. 20, 2013 to Office Action mailed May 29, 2013", (w/ English Translation of Amended Claims), 5 pgs.
"Japanese Application Serial No. 2009-549102, Response filed Mar. 22, 2013 to Office Action mailed Oct. 16, 2012", (w/ English Translation of Amended Claims), 7 pgs.
"Japanese Application Serial No. 2009-549102, Voluntary Amendment filed Feb. 7, 2011", (w/ English Translation of Amended Claims), 24 pgs.
"Japanese Patent Application Serial No. 2008-528017, Office Action mailed May 22, 2012", (English Translation), 4 pgs.
"Mexican Application Serial No. MX/a/2010/008697, Office Action mailed Nov. 28, 2011", 4 pgs.
"Mexican Application Serial No. MX/a/2010/008697, Response filed Jul. 10, 2012 to Office Action mailed May 4, 2012", 5 pgs.
"Mexican Application Serial No. MX/a/2010/8697, Office Action mailed May 10, 2012", (English Translation), 3 pgs.
"Mexican Appplication Serial No. MX/a/2010/008697 , Office Action Response Filed Mar. 28, 2012", 22 pgs.
"Singapore Applicaiton Serial No. 201005638-0, Office Action mailed Jun. 27, 2012", 7 pgs.
"Singapore Application Serial No. 201005638-0, Office Action mailed Nov. 9, 2011", 16 pgs.
"Singapore Application Serial No. 201005638-0, Office Action Response filed Mar. 29, 2012 to Office Action mailed Nov. 9, 2011", (English Translation), 91 pgs.
"Singapore Application Serial No. 201005638-0, Response filed Aug. 22, 2012 to Office Action mailed Jun. 27, 2012", 2 pgs.
"Singapore Application Serial No. 201005638-0, Search Report mailed Oct. 27, 2011", 7 pgs.
"Singapore Application Serial No. 201005638-0, Written Opinion mailed Oct. 27, 2011", 8 pgs.
"U.S. Appl. No. 12/027,960, Amendment Under 37 C.F.R. Sec. 1.312 filed Nov. 1, 2012", 7 pgs.
"U.S. Appl. No. 12/027,960, Non Final Office Action mailed Apr. 10, 2012", 16 pgs.
"U.S. Appl. No. 12/027,960, Notice of Allowance mailed Aug. 1, 2012", 11 pgs.
"U.S. Appl. No. 12/027,960, Preliminary Amendment mailed Dec. 8, 2010", 21 pgs.
"U.S. Appl. No. 12/027,960, Response filed Jul. 10, 2012 to Non Final Office Action mailed Apr. 10, 2012", 8 pgs.
"U.S. Appl. No. 12/027,960, Response filed Oct. 24, 2011 to Restriction Requirement mailed Sep. 23, 2011", 21 pgs.
"U.S. Appl. No. 12/027,960, Response to Rule 312 Communication mailed Nov. 14, 2012", 2 pgs.
"U.S. Appl. No. 12/027,960, Restriction Requirement mailed Sep. 23, 2011", 9 pgs.
"U.S. Appl. No. 12/064,529 , Response filed Jul. 9, 2012 to Non Final Office Action mailed Apr. 9, 2012", 11 pgs.
"U.S. Appl. No. 12/064,529, Final Office Action mailed Sep. 20, 2012", 14 pgs.
"U.S. Appl. No. 12/064,529, Non Final Office Action mailed Apr. 9, 2012", 15 pgs.
"U.S. Appl. No. 12/064,529, Preliminary Amendment filed Feb. 22, 2008", 11 pgs.
"U.S. Appl. No. 12/064,529, Response filed Oct. 24, 2011 to Restriction Requirement mailed Aug. 24, 2011", 9 pgs.
"U.S. Appl. No. 12/064,529, Restriction Requirement mailed Aug. 24, 2011", 9 pgs.
"U.S. Appl. No. 12/302,738, Final Office Action mailed Oct. 3, 2013", 11 pgs.
"U.S. Appl. No. 12/302,738, Non Final Office Action mailed Jan. 2, 2013", 10 pgs.
"U.S. Appl. No. 12/302,738, Notice of Allowance mailed Dec. 27, 2013", 9 pgs.
"U.S. Appl. No. 12/302,738, Preliminary Amendment filed Nov. 26, 2008", 8 pgs.
"U.S. Appl. No. 12/302,738, Response filed Dec. 3, 2013 to Final Office Action mailed Oct. 3, 2013", 8 pgs.
"U.S. Appl. No. 12/302,738, Response filed Jun. 26, 2013 to Non Final Office Action mailed Jan. 2, 2013", 10 pgs.
"U.S. Appl. No. 12/302,738, Response filed Nov. 19, 2012 to Restriction Requirement mailed Oct. 19, 2012", pgs.
"U.S. Appl. No. 12/302,738, Restriction Requirement mailed Oct. 19, 2012", 7 pgs.
"U.S. Appl. No. 12/367,172 , Response filed Aug. 13, 2012 to Final Office Action mailed Apr. 13, 2012", 9 pgs.
"U.S. Appl. No. 12/367,172, Final Office Action mailed Apr. 13, 2012", 21 pgs.
"U.S. Appl. No. 12/367,172, Final Office Action mailed Jan. 18, 2012", 15 pgs.
"U.S. Appl. No. 12/367,172, Non Final Office Action mailed May 27, 2011", 20 pgs.
"U.S. Appl. No. 12/367,172, Response filed Mar. 8, 2011 to Restriction Requirement mailed Dec. 8, 2010", 11 pgs.
"U.S. Appl. No. 12/367,172, Response filed Nov. 16, 2011 to Non Final Office Action mailed May 27, 2011", 6 pgs.
"U.S. Appl. No. 12/367,172, Restriction Requirement mailed Dec. 8, 2010", 6 pgs.
"U.S. Appl. No. 12/704,343, Advisory Action mailed Apr. 5, 2013", 3 pgs.
"U.S. Appl. No. 12/704,343, Examiner Interview Summary mailed Feb. 25, 2013", 3 pgs.
"U.S. Appl. No. 12/704,343, Examiner Interview Summary mailed Feb. 7, 2013", 3 pgs.
"U.S. Appl. No. 12/704,343, Final Office Action mailed Dec. 7, 2012", 9 pgs.
"U.S. Appl. No. 12/704,343, Final Office Action mailed May 10, 2013", 7 pgs.
"U.S. Appl. No. 12/704,343, Non Final Office Action mailed Jul. 16, 2012", 14 pgs.
"U.S. Appl. No. 12/704,343, Notice of Allowance mailed Aug. 2, 2013", 10 pgs.
"U.S. Appl. No. 12/704,343, Notice of Allowance mailed Jan. 3, 2014", 11 pgs.
"U.S. Appl. No. 12/704,343, Response filed Feb. 27, 2013 to Final Office Action mailed Dec. 7, 2012", 9 pgs.
"U.S. Appl. No. 12/704,343, Response filed Jul. 10, 2013 to Final Office Action mailed May 10, 2013", 8 pgs.
"U.S. Appl. No. 12/704,343, Response filed Jul. 5, 2012 to Restriction Requirement mailed May 7, 2012", 7 pgs.
"U.S. Appl. No. 12/704,343, Response filed Oct. 16, 2 to Non Final Office Action mailed Jul. 16, 2012", 12 pgs.
"U.S. Appl. No. 12/704,343, Restriction Requirement mailed May 7, 2012", 7 pgs.
"U.S. Appl. No. 13/736,545, Notice of Allowance mailed Aug. 2, 2013", 9 pgs.
"U.S. Appl. No. 13/736,545, Preliminary Amendment filed Mar. 6, 2013", 3 pgs.
"U.S. Appl. No. 13/791,175, Final Office Action mailed Dec. 26, 2013", 12 pgs.
"U.S. Appl. No. 13/791,175, Non Final Office Action mailed Jun. 7, 2013", 10 pgs.
"U.S. Appl. No. 13/791,175, Preliminary Amendment filed Mar. 8, 2013", 4 pgs.
"U.S. Appl. No. 13/791,175, Response filed Nov. 1, 2013 to Non Final Office Action mailed Jun. 7, 2013", 8 pgs.
Anders, H.-J., et al., "Molecular mechanisms of autoimmunity triggered by microbial infection", Arthritis Research & Therapy, 7(5), (2005), 215-224.
Australian Application Serial No. 2008227128, Secondary Amendment filed Jan. 9, 2012, 16 pgs.
Baenziger, S., et al., "Triggering TLR7 in mice induces immune activation and lymphoid system disruption, resembling HIV-mediated pathology", Blood, 113(2), (Jan. 8, 2009), 377-388.
Brown, Gordon. Dectin-1: a signalling non-TLR pattern-recognition receptor. Nature Reviews Immunology 2006. 6: 33-43. *
Bryan, G. T., et al., "Interferon (IFN) and IFN Inducers Protect Mouse Bladder Urothelium Against Carcinogenicity by FANFT", Journal of Cancer Research and Clinical Oncology, 116(Suppl. Part 1), (Abstract A3.106.36), (15th International Cancer Congress, Hamburg, Aug. 16-22, 1990), (1990), p. 308.
Butler, Roslyn S, et al., "Highly fluorescent donor-acceptor purines", J. Mater. Chem., 17(19), (2007), 1863-1865.
Canadian Application Serial No. 2,677,733 Response filed Feb. 16, 2015 to Office Action mailed Aug. 25, 2014, 6 pgs.
Canadian Application Serial No. 2,677,733, Office Action mailed Aug. 25, 2014, 2 pgs.
Carson, D. A., et al., "TLR Agonists", U.S. Appl. No. 60/710,337, filed Aug. 22, 2005, 52 pgs.
Chan, M., et al., "Synthesis and immunological characterization of toll-like receptor 7 agonistic conjugates", Bioconjug Chem., 20(6), (Jun. 2009), 1194-200.
Colombo, R., et al., "Combination of intravesical chemotherapy and hyperthermia for the treatment of superficial bladder cancer: preliminary clinical experience", Crit Rev Oncol Hematol., 47(2), (Aug. 2003), 127-39.
Dolan, M. E, et al., "Metabolism of O6-benzylguanine, an inactivator of O6-alkylguanine-DNA alkyltransferase.", Cancer Res., 54(19), (Oct. 1, 1994), 5123-30.
European Application Serial No. 06813535.9, Response filed Apr. 4, 2014 to Examination Notification Art. 94(3) mailed Sep. 24, 2013, 70 pgs.
European Application Serial No. 07755916.9, Office Action mailed Mar. 25, 2014, 1 pg.
European Application Serial No. 07755916.9, Response filed May 23, 2014 to Examination Notification Art. 94(3) mailed Aug. 15, 2013, 13 pgs.
European Application Serial No. 12004181.9 Response Filed Dec. 15, 2014 to Non-Final Office Action Mailed Jul. 18, 2014, 138 pgs.
European Application Serial No. 12004181.9, Examination Notification Art. 94(3) mailed Jul. 18, 2014, 4 pgs.
European Application Serial No. 12004181.9, Response filed Mar. 7, 2014 to Examination Notification Art. 94(3) mailed Sep. 2, 2013, 10 pgs.
European Application Serial No. 13001957.3, Extended European Search Report mailed Jan. 28, 2014, 17 pgs.
European Application Serial No. 13001957.3, Office Action mailed Mar. 3, 2014, 2 pgs.
European Application Serial No. 13001957.3, Response filed Aug. 20, 2014 Extended European Search Report mailed Jan. 28, 2014, Includes Response Office Action mailed Mar. 3, 2014, 7 pgs.
Hayashi, T., et al., "Mast cell-dependent anorexia and hypothermia induced by mucosal activation of Toll-like receptor 7", Am J Physiol Regul Integr Comp Physiol., 295(1), (2008), R123-32.
Horner et al., Optimized conjugation ratios lead to allergen immunostimulatory oligodeoxynucleotide conjugates with retained immunogenicity and minimal anaphylactogenicity. J Allergy Clin Immunol, 2002;110:413-20. *
Indian Application Serial No. 5675/DELNP/2009, First Examiner Report mailed Sep. 11, 2014, 2 pgs.
Interchim Inc., Isolation/Modification/Labeling Product Sheet, Jan. 15, 2000, p. 1-23. *
Jacobson, Kenneth A, et al., "Adenosine analogs with covalently attached lipids have enhanced potency at Al-adenosine receptors", FEBS Letters, 225(1-2), (1987), 97-102.
Japanese Application Serial No. 2013-59721, Office Action mailed May 30, 2014, w/English translation, 10 pgs.
Jin, "", Bioorganic & Medicianl Chemistry Letter, vol. 16, No. 17, (2006), 4559-4563.
Jin, G., et al., "Synthesis and immunostimulatory activity of 8-substituted amino 9-benzyladenines as potent Toll-like receptor 7 agonists.", Bioorg Med Chem Lett., 16(17), (Sep. 1, 2006), 4559-63.
Julien ("Chapter 2: Pharmacodynamics: How Drugs Act", A Primer of Drug Action (Ninth Edition); Worth Publishers, 2001:37-57. *
Kobayashi, H., et al., "Prepriming: a novel approach to DNA-based vaccination and immunomodulation", Springer Seminars in Immunopathology, 22(Nos. 1-2), (2000), 85-96.
Korean Application Serial No. 10-2009-7018499, Office Action mailed Sep. 17, 2014, 8 pgs.
Kulikov, V. I, et al., "Lipid derivatives of prostaglandins and nonsteroidal antiinflammatory drugs (a review)", Pharmaceutical Chemistry Journal, 31(4), (1997), 173-177.
Kurimoto, A., et al., "Synthesis and evaluation of 2-substituted 8-hydroxyadenines as potent interferon inducers with improved oral bioavailabilities", Bioorg Med Chem., 12(5), (Mar. 1, 2004), 1091-9.
Kurimoto, A., et al., "Synthesis and structure-activity relationships of 2-amino-8-hydroxyadenines as orally active interferon inducing agents", Bioorg Med Chem., 11(24), (Dec. 1, 2003), 5501-8.
Lee, J., et al., "Molecular basis for the immunostimulatory activity of guanine nucleoside analogs: Activation of Toll-like receptor 7", Proc. Natl. Acad. Sci., 100(11), (2003), 6646-6651.
Lippard, Stephen J. The Art of Chemistry. Nature 2002. 416, p. 587. *
Liu, H., et al., "Tumour growth inhibition by an imidazoquinoline is associated with c-Myc down-regulation in urothelial cell carcinoma", BJU International, 101(7), (Apr. 2008), 894-901.
Mayer, R., et al., "A randomized controlled trial of intravesical bacillus calmette-guerin for treatment refractory interstitial cystitis", Journal of Urology, 173(4), (Apr. 2005), 1186-1191.
Metzler, David E, "Biosynthesis of triglycerides and phospholipids", Biochemistry: The Chemical Reactions of Living Cells, (1977), 3 pgs.
Miller, R L, et al., "Imiquimod applied topically: a novel immune response modifier and new class of drug", Int J Immunopharmacol., 21(1), (Jan. 1999), 1-14.
Mosmann, T. R., et al., "TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties", Annual Review Immunology, 7, (1989), 145-173.
Rohn, S., et al., "Antioxidant activity of protein-bound quercetin", J Agric Food Chem., 52(15), (Jul. 28, 2004), 4725-9.
Schon, M., et al., "Tumor-Selective Induction of Apoptosis and the Small-Molecule Immune Response Modifier Imiquimod", J Natl Cancer Inst, 95(15), (2003), 1138-1149.
Sidky, Y. A, et al., "Effects of Treatment with an Oral Interferon Inducer, Imidazoquinolinamine (R-837), on the Growth of Mouse Bladder Carcinoma FCB", Journal of Interferon Research, 10(Supp 1), (Abstract II6-12) (Annual Meeting of the ISIR, San Francisco, CA, Nov. 14-18, 1990), (Nov. 1990), S123.
Sidky, Y. A, et al., "Inhibition of murine tumor growth by an interferon-inducing imidazoquinolinamine", Cancer Research, 52(13), (Jul. 1, 1992), 3528-33.
Sidky, Y. A., et al., "Curative effectiveness of the interferon inducing imiquimod as a signal agent in mouse bladder tumors", Proceedings, Eighty-Fourth Meeting of the American Association for Cancer Research, vol. 34, (Abstract 2789) (May 19-22, 1993, Orlando, FL), (Mar. 1993), p. 467.
Sidky, Y. A., et al., "Effects of treatment with the oral interferon inducer, R-837, on the growth of mouse colon carcinoma, MC-26", Proceedings, 81st Annual Meeting of the American Association for Cancer Research, vol. 31, (Abstract 2574), (Mar. 1990), p. 433.
Sidky, Y. A., et al., "Inhibition of tumor-induced angiogenesis by the interferon inducer Imiquimod", Proceedings, Eighty-Third Annual Meeting of the American Association of Cancer Research, vol. 33, (Abstract 458) (May 20-23, 1992, San Diego, CA), (Mar. 1992), p. 77.
Simons, M. P., et al., "Identification of the Mycobacterial Subcomponents Involved in the Release of Tumor Necrosis Factor Related Apoptosis-Including Ligand from Human Neutrophills Infection and Immunity", Infection and Immunity, 75(3), (2007), 1265-1271.
Simons, M. P., et al., "Identification of the Mycobacterial Subcomponents Involved in the Release of Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand from Human Neutrophils", Infection and Immunity, 75(3), (2007), 1265-1271.
Smith, E. B, et al., "Antitumor effects of imidazoquinolines in urothelial cell carcinoma of the bladder", J Urol., 177(6), (May 2007), 2347-51.
Smith, E. B., et al., "Antitumor Effects of Imidazoquinolines in Urothelial Cell Carcinoma of the Bladder", The Journal of Urology, 177(6), (Abstract Only), (2007), 3 pgs.
Smith, E. B., et al., "Effects of Imiquimod, a toll-like receptor-7 agonist, on cell proliferation and cytokine production in bladder cancer in vitro and in vivo", Journal of Urology, 173(4, Suppl. S), (Apr. 2005), p. 158.
Smith, Eric B., et al., "Antitumor Effects of Imidazoquinolines in Urothelial Cell Carcinoma of the Bladder", The Journal of Urology, 177(6), (Jun. 2007), 2347-2351.
Spohn, R., et al., "Synthetic lipopeptide adjuvants and Toll-like receptor 2-structure-activity relationships", Vaccine, 22(19), (Jun. 23, 2004), 2494-9.
Staros, E. B., et al., "New Approaches to Understanding Its Clinical Significance", Am. J. Clin. Pathol., 123(2) (2005), 305-312.
Takeda, K., et al., "Toll-like receptors in innate immunity", International Immunology, 17(1), (2005), 1-14.
Takeda, K., et al., "Toll-like receptors", Annu Rev Immunol., 21, (2003), 335-76.
U.S. Appl. No. 12/302,738, Notice of Allowance mailed Apr. 22, 2014, 7 pgs.
U.S. Appl. No. 12/367,172, Final Office Action mailed Apr. 21, 2015, 19 pgs.
U.S. Appl. No. 12/367,172, Non Final Office Action mailed Jul. 1, 2014, 20 pgs.
U.S. Appl. No. 12/367,172, Response filed Dec. 29, 2014 to Non Final Office Action mailed Jul. 1, 2014, 7 pgs.
U.S. Appl. No. 13/736,545, Notice of Allowance mailed Mar. 18, 2014, 6 pgs.
U.S. Appl. No. 13/791,175, Final Office Action mailed Nov. 20, 2014, 12 pgs.
U.S. Appl. No. 13/791,175, Non Final Office Action mailed Jul. 21, 2014, 11 pgs.
U.S. Appl. No. 13/791,175, Response filed Jun. 26, 2014 to Final Office Action mailed Dec. 26, 2013, 9 pgs.
U.S. Appl. No. 13/791,175, Response filed Oct. 21, 2014 to Non Final Office Action mailed Jul. 21, 2014, 7 pgs.
U.S. Appl. No. 14/309,245, Notice of Allowance mailed Jan. 20, 2015, 12 pgs.
Veronese, F. M., et al., "The impact of PEGylation on biological therapies", BioDrugs, 22(5), (2008), 315-329.
Wille-Reece, U., et al., "HIV Gag protein conjugated to a Toll-like receptor 7/8 agonist improves the magnitude and quality of Th1 and CD8+ T cell responses in nonhuman primates", Proc. Natl. Acad. Sci. USA, 102(42), (Oct. 18, 2005), 15190-15194.
Wille-Reece, Ulrike, "", PNAS, vol. 102, No. 42, (2005), 15190-15194.
Wu, C., et al., "Immunotherapeutic activity of a conjugate of a Toll-like receptor 7 ligand", Proc. Natl. Acad. Sci. USA, 104(10), (2007), 3990-3995.
Wu, Christina C N, "", PNAS, vol. 104, No. 10, (Mar. 6, 2007), 3990-3995.
Yang, Victor C., et al., "Bioconjugates for Effective Drug Targeting", Advanced Drug Delivery Reviews 55 (2003), (2002), 169-170.
Zaks, K, et al., "Efficient immunization and cross-priming by vaccine adjuvants containing TLR3 or TLR9 agoinst complexed to cationic Liposomes", Journal of Immunology, 176(12), (Jun. 15, 2006), 7335-7345.
Zaks, Karen, "", The Journal of Immunology, vol. 176, No. 12, (2006), 7335-7345.

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11697851B2 (en) 2016-05-24 2023-07-11 The Regents Of The University Of California Early ovarian cancer detection diagnostic test based on mRNA isoforms
EP4252629A2 (en) 2016-12-07 2023-10-04 Biora Therapeutics, Inc. Gastrointestinal tract detection methods, devices and systems
WO2018112223A1 (en) 2016-12-14 2018-06-21 Progenity Inc. Treatment of a disease of the gastrointestinal tract with a tlr modulator
US11426566B2 (en) 2016-12-14 2022-08-30 Biora Therapeutics, Inc. Treatment of a disease of the gastrointestinal tract with a TLR modulator
WO2021174024A1 (en) 2020-02-28 2021-09-02 First Wave Bio, Inc. Methods of treating iatrogenic autoimmune colitis
WO2022003598A1 (en) 2020-07-02 2022-01-06 Viiv Healthcare Company Method of achieving hiv viral remission using long-acting antiretroviral agents

Also Published As

Publication number Publication date
EP1931352A4 (en) 2011-11-23
JP2013040209A (en) 2013-02-28
WO2007024707A3 (en) 2007-09-20
EP1931352B1 (en) 2016-04-13
AU2006283524A2 (en) 2008-06-05
US20090324551A1 (en) 2009-12-31
CA2620182A1 (en) 2007-03-01
CN101304748A (en) 2008-11-12
AU2006283524A1 (en) 2007-03-01
JP2009504803A (en) 2009-02-05
US20130165455A1 (en) 2013-06-27
WO2007024707A2 (en) 2007-03-01
ES2577514T3 (en) 2016-07-15
EP1931352A2 (en) 2008-06-18

Similar Documents

Publication Publication Date Title
US9359360B2 (en) TLR agonists
ES2456964T3 (en) Conjugates of synthetic TLR agonists and their uses
US8846697B2 (en) Purine analogs
US9441005B2 (en) Toll-like receptor-7 and -8 modulatory 1H imidazoquinoline derived compounds
AU2006283524B2 (en) TLR agonists

Legal Events

Date Code Title Description
AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:UNIVERSITY OF CALIFORNIA SAN DIEGO;REEL/FRAME:029355/0995

Effective date: 20121126

AS Assignment

Owner name: THE REGENTS OF THE UNIVERSITY OF CALIFORNIA, CALIF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CARSON, DENNIS A;GRIMSHAW, SUZANNE;COTTAM, HOWARD B.;AND OTHERS;SIGNING DATES FROM 20080626 TO 20080627;REEL/FRAME:031569/0593

FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4

CC Certificate of correction
MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 8